WO1994013300A1 - Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies - Google Patents

Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies Download PDF

Info

Publication number
WO1994013300A1
WO1994013300A1 PCT/US1993/011857 US9311857W WO9413300A1 WO 1994013300 A1 WO1994013300 A1 WO 1994013300A1 US 9311857 W US9311857 W US 9311857W WO 9413300 A1 WO9413300 A1 WO 9413300A1
Authority
WO
WIPO (PCT)
Prior art keywords
metal complex
salen
group
antioxidant salen
antioxidant
Prior art date
Application number
PCT/US1993/011857
Other languages
English (en)
Other versions
WO1994013300A9 (fr
Inventor
Bernard Malfroy-Camine
Michel Baudry
Original Assignee
Eukarion, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU57419/94A priority Critical patent/AU697399B2/en
Priority to GB9415050A priority patent/GB2277873B/en
Priority to KR10-2003-7004595A priority patent/KR20040004386A/ko
Priority to UA95062707A priority patent/UA27949C2/uk
Priority to CA002150937A priority patent/CA2150937C/fr
Priority to SK745-95A priority patent/SK74595A3/sk
Priority to EP94903498A priority patent/EP0746321B1/fr
Priority to HU9501644A priority patent/HU225109B1/hu
Priority to DE69333775T priority patent/DE69333775T2/de
Priority to JP51432894A priority patent/JP3502099B2/ja
Application filed by Eukarion, Inc. filed Critical Eukarion, Inc.
Priority to AT94903498T priority patent/ATE290867T1/de
Priority to PL93309334A priority patent/PL175446B1/pl
Publication of WO1994013300A1 publication Critical patent/WO1994013300A1/fr
Publication of WO1994013300A9 publication Critical patent/WO1994013300A9/fr
Priority to LVP-95-158A priority patent/LV10924B/en
Priority to NO19952237A priority patent/NO319454B1/no
Priority to US08/479,697 priority patent/US5834509A/en
Priority to US08/485,489 priority patent/US5696109A/en
Priority to US09/542,182 priority patent/US6573257B2/en
Priority to US10/452,695 priority patent/US6900198B2/en
Priority to US11/362,454 priority patent/US7582786B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F13/00Compounds containing elements of Groups 7 or 17 of the Periodic System
    • C07F13/005Compounds without a metal-carbon linkage

Definitions

  • the invention provides pharmaceutical compositions of synthetic catalytic small molecule antioxidants and free radical scavengers for therapy and prophylaxis of disease, methods for using the small molecule antioxidants in prevention and treatment of pathological conditions, methods for using the small molecule antioxidants for targeted protection of tissues and/or cell types during cancer chemotherapy, and methods for using the small molecule antioxidants to prevent toxicologic damage to individuals exposed to irritating oxidants or other sources of oxidative damage, particularly oxygen-derived oxidative species such as superoxide radical.
  • the compositions and methods of the invention are also used for preventing oxidative damage in human transplant organs and for inhibiting reoxygenation injury following reperfusion of ischemic tissues.
  • the compositions and methods of the invention are also useful for chemoprevention of chemical carcinogenesis and alteration of drug metabolism involving epoxide or free oxygen radical intermediates.
  • Oxygen is an essential nutrient for nonfacultative aerobic organisms, including, of course, humans.
  • Oxygen is used in many important ways, namely, as the terminal electronic acceptor in oxidative phosphorylation, in many dioxygenase reactions, including the synthesis of prostaglandins and of vitamin A from carotenoids, in a host of hydroxylase reactions, including the formation and modification of steroid hormones, and in both the activation and the inactivation of xenobiotic ⁇ , including carcinogens.
  • the extensive P-450 system uses molecular oxygen in a host of important cellular reactions. In a similar vein, nature employs free radicals in a large variety of enzymic reactions.
  • Biological antioxidants include well-defined enzymes, such as superoxide dismutase, catalase, selenium glutathione peroxidase, and phospholipid hydroperoxide glutathione peroxidase.
  • Nonenzymatic biological antioxidants include tocopherols and tocotrienols, carotenoids, quinones, bilirubin, ascorbic acid, uric acid, and metal-binding proteins.
  • Various antioxidants being both lipid and water soluble, are found in all parts of cells and tissues, although each specific antioxidant often shows a characteristic distribution pattern.
  • the so-called ovothiols which are ercaptohistidine derivatives, also decompose peroxides nonenzymatically.
  • Oxyradical injury has been implicated in the pathogenesis of pulmonary oxygen toxicity, adult respiratory distress syndrome (ARDS) , bronchopulmonary dysplasia, sepsis syndrome, and a variety of ischemia-reperfusion syndromes, including myocardial infarction, stroke, cardiopulmonary bypass, organ transplantation, necrotizing enterocolitis, acute renal tubular necrosis, and other disease.
  • ARDS adult respiratory distress syndrome
  • ischemia-reperfusion syndromes including myocardial infarction, stroke, cardiopulmonary bypass, organ transplantation, necrotizing enterocolitis, acute renal tubular necrosis, and other disease.
  • Oxyradicals can react with proteins, nucleic acids, lipids, and other biological macromolecules producing damage to cells and tissues, particularly in the critically ill patient.
  • Free radicals are atoms, ions, or molecules that contain an unpaired electron (Pryor, A (1976) Free Radicals in Biol. 1 : 1) . Free radicals are usually unstable and exhibit short half-lives. Elemental oxygen is highly electronegative and readily accepts single electron transfers from cytochromes and other reduced cellular components; a portion of the 0 2 consumed by cells engaged in aerobic respiration is univalently reduced to superoxide radical (•0 2 ⁇ ) (Cadenas E (1989) Ann. Rev. Biochem. 58: 79) . Sequential univalent reduction of -0 2 ⁇ produces hydrogen peroxide (H 2 0 2 ) , hydroxyl radical (»OH) , and water.
  • Free radicals can originate from many sources, including aerobic respiration, cytochrome P-450-catalyzed monooxygenation reactions of drugs and xenobiotics (e.g., trichloromethyl radicals, CC1 3 » formed from oxidation of carbon tetrachloride) , and ionizing radiation.
  • cytochrome P-450-catalyzed monooxygenation reactions of drugs and xenobiotics e.g., trichloromethyl radicals, CC1 3 » formed from oxidation of carbon tetrachloride
  • ionizing radiation e.g., when tissues are exposed to gamma radiation, most of the energy deposited in the cells is absorbed by water and results in scission of the oxygen-hydrogen covalent bonds in water, leaving a single electron on hydrogen and one on oxygen creating two radicals H « and «OH.
  • the hydroxyl radical, »OH is the most reactive radical known in chemistry.
  • superoxide may be produced physiologically by endothelial cells for reaction with nitric oxide, a physiological regulator, forming peroxynitrite, ONOO " which may decay and give rise to hydroxyl radical, »OH (Marietta MA (1989) Trends Biochem. Sci. 14: 488; Moncada et al. (1989) Biochem. Pharmacol. 38: 1709; Saran et al. (1990) Free Rad. Res. Commun. 10: 221; Beckman et al. (1990) Proc. Natl. Acad. Sci. fU.S.A.I 87: 1620) .
  • Additional sources of oxyradicals are "leakage" of electrons from disrupted mitochondrial or endoplasmic reticular electron transport chains, prostaglandin synthesis, oxidation of catecholamines, and platelet activation.
  • Many free radical reactions are highly damaging to cellular components; they crosslink proteins, mutagenize DNA, and peroxidize lipids. Once formed, free radicals can interact to produce other free radicals and non-radical oxidants such as singlet oxygen ( 1 02) and peroxides. Degradation of some of the products of free radical reactions can also generate potentially damaging chemical species.
  • malondialdehyde is a reaction product of peroxidized lipids that reacts with virtually any amine-containing molecule. Oxygen free radicals also cause oxidative modification of proteins (Stadtman ER (1992) Science 257: 1220) .
  • Aerobic cells generally contain a number of defenses against the deleterious effects of oxyradicals and their reaction products.
  • Superoxide dis utases catalyze the reaction:
  • H 2 o0 2 " + 2 H + > 0 2 + H 2 0 2 which removes superoxide and forms hydrogen peroxide.
  • H 2 0 2 is not a radical, but it is toxic to cells; it is removed by the enzymatic activities of catalase and glutathione peroxidase (GSH-Px) .
  • Catalase catalyzes the reaction:
  • GSH-Px removes hydrogen peroxide by using it to oxidize reduced glutathione (GSH) into oxidized glutathione (GSSG) according to the following reaction: 2 GSH + H 2 0 2 > GSSG + 2 H 2 0
  • PLOOH-GSH-Px phospholipid hydroperoxide glutathione peroxidase
  • PLOOH-GSH-Px phospholipid hydroperoxide glutathione peroxidase
  • Other enzymes such as phospholipid hydroperoxide glutathione peroxidase (PLOOH-GSH-Px) , converts reactive phospholipid hydroperoxides, free fatty acid hydroperoxides, and cholesterol hydroperoxides to corresponding harmless fatty acid alcohols.
  • Glutathione S-transferases also participate in detoxifying organic peroxides. In the absence of these enzymes and in presence of transition metals, such as iron or copper, superoxide and hydrogen peroxide can participate in the following reactions which generate the extremely reactive hydroxyl radical »0H " :
  • Carotenoids dramatically reduce the incidence of certain premalignant conditions, such as leukoplakia, in some patients.
  • a variety of antioxidants have been used.
  • Iron ion chelators such as desferrioxamine (also called deferoxamine or Desferol) and others, inhibit iron ion- dependent «0H generation and thus act as inhibitors of free radical formation (Gutteridge et al. (1979) Biochem. J. 184: 469; Halliwell B (1989) Free Radical Biol. Med. 7: 645; Van der Kraaij et al. (1989) Circulation 80: 158) .
  • Amino-steroid- based antioxidants such as the 21-aminosteroids termed "lazaroids" (e.g., U74006F) have also been proposed as inhibitors of oxyradical formation.
  • lazaroids e.g., U74006F
  • Desferrioxamine, allopurinol, and other pyrazolopyrimidines such as oxypurinol
  • oxypurinol have also been tested for preventing oxyradical formation in a myocardial stunning model system (Bolli et al. (1989) Circ. Res. 65: 607) and following hemorrhagic and endotoxic shock (DeGaravilla et al. (1992) Drug Devel. Res. 25: 139) .
  • each of these compounds has notable drawbacks for therapeutic usage.
  • deferoxamine is not an ideal iron chelator and its cellular penetration is quite limited.
  • Another strategy for preventing oxyradical-induced damage is to catalytically remove oxyradicals such as superoxide once they have been formed.
  • Superoxide dismutase and catalase have been extensively explored, with some success, as protective agents when added to reperfusates in many types of experiments or when added pre-ischemia (reviewed in Gutteridge JMC and Halliwell B (1990) op.cit. ) .
  • the availability of recombinant superoxide dismutase has allowed more extensive evaluation of the effect of administering SOD in the treatment or prevention of various medical conditions including reperfusion injury of the brain and spinal cord
  • An alternative strategy for preventing oxyradical- induced damage is to scavenge oxyradicals such as superoxide once these have been formed, typically by employing small molecule scavengers which act stoichiometrically rather than catalytically.
  • Congeners of glutathione have been used in various animal models to attenuate oxyradical injury.
  • N-2-mercaptopropionylglycine has been found to confer protective effects in a canine model of myocardial ischemia and reperfusion (Mitsos et al.
  • Mannitol has also been used as a free radical scavenger to reduce organ injury during reoxygenation (Fox RB (1984) J. Clin. Invest. 74: 1456; Ouriel et al. (1985) Circulation 72: 254) .
  • a small molecule chelate was reported to have activity as a glutathione peroxidase mimic (Spector et al. (1993) Proc. Natl. Acad. Sci. (U.S.A.) 90: 7485) .
  • inhibitors of oxyradical formation and/or enzymes that remove superoxide and hydrogen peroxide and/or small molecule oxyradical scavengers have all shown promise for preventing reoxygenation damage present in a variety of ischemic pathological states and for treating or preventing various disease states associated with free radicals.
  • each of these categories contains several drawbacks.
  • inhibitors of oxyradical formation typically chelate transition metals which are used in essential enzymatic processes in normal physiology and respiration; moreover, even at very high doses, these inhibitors do not completely prevent oxyradical formation.
  • Superoxide dismutases and catalase are large polypeptides which are expensive to manufacture, do not penetrate cells or the blood-brain barrier, and generally require parenteral routes of administration. Free radical scavengers act stoichiometrically and are thus easily depleted and must be administered in high dosages to be effective. Based on the foregoing, it is clear that a need exists for antioxidant agents which are efficient at removing dangerous oxyradicals, particularly superoxide and hydrogen peroxide, and which are inexpensive to manufacture, stable, and possess advantageous pharmacokinetic properties, such as the ability to cross the blood-brain barrier and penetrate tissues. Such versatile antioxidants would find use as pharmaceuticals, chemoprotectants, and possibly as dietary supplements.
  • compositions which have potent antioxidant and/or free radical scavenging properties and function as in vivo antioxidants.
  • the pharmaceutical compositions of the invention comprise an efficacious dosage of at least one species of salen-transition metal complex, typically a salen-manganese complex such as a salen-Mn(III) complex.
  • the pharmaceutical composition comprises a salen-Mn complex which is a chelate of Mn(III) with a diamine derivative, such as ethylenediamine linked to two substituted salicylaldehydes.
  • compositions possess the activity of dismutating superoxide (i.e., superoxide dismutase activity) and, advantageously, also converting hydrogen peroxide to water (i.e., catalase activity).
  • the pharmaceutical compositions are effective at reducing pathological damage related to formation of oxyradicals such as superoxide and peroxides and other free radical species.
  • the invention also provides methods for treating and preventing pathological conditions by applying or administering compositions of salen-transition metal complexes in a therapeutic or prophylactic dosage.
  • Salen-transition metal complexes used in the methods of the invention are typically salen-manganese complexes, such as Mn(III)-salen complexes.
  • the invention provides methods for preventing or reducing ischemic/reperfusion damage to critical tissues such as the myocardium and central nervous system.
  • the invention also provides methods for preventing or reducing cellular damage resulting from exposure to various chemical compounds which produce potentially damaging free radical species, comprising administering a therapeutically or prophylactically efficacious dosage of at least one species of salen-transition metal complex, preferably a salen-manganese complex having detectable SOD activity and preferably also having detectable catalase activity.
  • the antioxidant salen-transition metal complexes of the invention are administered by a variety of routes, including parenterally, topically, and orally.
  • a therapeutic or prophylactic dosage of a salen-transition metal complex of the present invention is administered alone or combined with (1) one or more antioxidant enzymes, such as a Mn-SOD, a Cu,Zn- SOD, or catalase, and/or (2) one or more free radical scavengers, such as tocopherol, ascorbate, glutathione, DMTU, N-acetylcysteine, or N-2-mercaptopropionylglycine and/or (3) one or more oxyradical inhibitors, such as desferrioxamine or allopurinol, and/or one or more biological modifier agents, such as calpain inhibitors.
  • one or more antioxidant enzymes such as a Mn-SOD, a Cu,Zn- SOD, or catalase
  • free radical scavengers such as tocopherol, ascorbate, glutathione, DMTU, N-acetylcysteine, or N-2-mercaptopro
  • compositions of these compositions is dependent upon the specific pathological condition sought to be treated or prevented, the route and form of administration, and the age, sex, and condition of the patient.
  • These compositions are administered for various indications, including: (1) for preventing ischemic/reoxygenation injury in a patient, (2) for preserving organs for transplant in an anoxic, hypoxic, or hyperoxic state prior to transplant, (3) for protecting normal tissues from free radical-induced damage consequent to exposure to ionizing radiation and/or chemotherapy, as with bleomycin, (4) for protecting cells and tissues from free radical-induced injury consequent to exposure to xenobiotic compounds which form free radicals, either directly or as a consequence of monooxygenation through the cytochrome P-450 system, (5) for enhancing cryopreservation of cells, tissues, organs, and organisms by increasing viability of recovered specimens, and (6) for prophylactic administration to prevent: carcinogenesis, cellular senescence, cataract formation, formation of malondialdehyde adducts
  • salen-transition metal complexes are formulated for administration by the oral route by forming a pharmaceutical dosage form comprising an excipient and not less than 1 ⁇ g nor more than about 10 grams of at least one antioxidant salen-transition metal complex of the invention.
  • Dietary formulations are administered for therapy of free radical-induced diseases and/or for the chemoprevention of neoplasia and/or oxidative damage associated with normal aerobic metabolism.
  • buffered aqueous solutions comprising at least one antioxidant salen-transition metal complex of the invention at a concentration of at least 1 nM but not more than about 100 mM is formulated for administration, usually at a concentration of about 0.1 to 10 mM, typically by intravenous route, to a patient undergoing or expected to undergo: (1) an ischemic episode, such as a myocardial infarction, cerebral ischemic event, transplantation operation, open heart surgery, elective angioplasty, coronary artery bypass surgery, brain surgery, renal infarction, traumatic hemorrhage, tourniquet application, (2) antineoplastic or antihelminthic chemotherapy employing a chemotherapeutic agent which generates free radicals, (3) endotoxic shock or sepsis, (4) exposure to ionizing radiation, (5) exposure to exogenous chemical compounds which are free radicals or produce free radicals, (6) thermal or chemical burns or ulcerations, (7) hyperbaric oxygen, or (8) apoptosis
  • the buffered aqueous solutions of the invention may also be used, typically in conjunction with other established methods, for organ culture, cell culture, transplant organ maintenance, and myocardial irrigation.
  • Nonaqueous formulations such as lipid-based formulations are also provided, including stabilized emulsions.
  • the antioxidant salen-metal compositions are administered by various routes, including intravenous injection, intramuscular injection, subdermal injection, intrapericardial injection, surgical irrigation, topical application, ophthalmologic application, lavage, gavage, enema, intraperitoneal infusion, mist inhalation, oral rinse, and other routes, depending upon the specific medical or veterinary use intended.
  • antioxidant salen-transition metal complexes of the invention are employed to modulate the expression of naturally-occurring genes or other polynucleotide sequences under the transcriptional control of an oxidative stress response element (e.g., an antioxidant responsive element, ARE) , such as an antioxidant response element of a glutathione S-transferase gene or a NAD(P)H:quinone reductase gene.
  • ARE antioxidant responsive element
  • the antioxidant salen-metal complexes may be used to modulate the transcription of ARE- regulated polynucleotide sequences in cell cultures (e.g., ES cells) and in intact animals, particularly in transgenic animals wherein a transgene comprises one or more AREs as transcriptional regulatory sequences.
  • the present invention also encompasses pharmaceutical compositions of antioxidant salen-manganese complexes, therapeutic uses of such antioxidant salen- manganese complexes, methods and compositions for using antioxidant salen-manganese complexes in diagnostic, therapeutic, and research applications in human and veterinary medicine.
  • the invention also provides methods for preventing food spoilage and oxidation by applying to foodstuffs an effective amount of at least one antioxidant salen-metal complex species.
  • the invention also provides compositions for preventing food spoilage comprising an effective amount of at least one species of antioxidant salen-metal complex, optionally in combination with at least one additional food preservative agent (e.g., butylated hydroxytoluene,.
  • an antioxidant salen-metal complex is incorporated into a foodstuff subject to rancidification (e.g., oxidation) to reduce the rate of oxidative decomposition of the foodstuff when exposed to molecular oxygen.
  • Fig. 1 shows the general structure of salen deriviatives of the invention.
  • Fig. 2 shows a salen derivative according to the structure shown in Figure 1, wherein n is 0.
  • Fig. 3 shows structures of preferred compounds of the invention.
  • Fig. 4 shows schematically the effect of an ischemic/reoxygenation episode on synaptic transmission in isolated brain slices.
  • Fig. 5 shows the effect of a salen-Mn complex on EPSP amplitude following an episode of ischemia/reoxygenation.
  • Fig. 6 shows the effect of a salen-Mn complex on EPSP initial slope following an episode of ischemia/reoxygenation.
  • Fig. 7 shows the effect of a salen-Mn complex on brain slice viability following repeated episodes of ischemia/reoxygenation.
  • Fig. 8 shows the protective effect of a salen-Mn complex in an animals model of iatrogenic Parkinson's disease.
  • Fig. 9 shows that C7 protects hippocampal slices from lactic acid-induced lipid peroxidation.
  • Fig 10 shows C7 protects dopaminergic neurons in mouse striatum from 6-OHDA-induced degeneration.
  • Fig 11 shows C7 protects dopaminergic neurons in mouse striatum from MPTP-induced degeneration.
  • an "antioxidant” is a substance that, when present in a mixture or structure containing an oxidizable substrate biological molecule, significantly delays or prevents oxidation of the substrate biological molecule.
  • Antioxidants can act by scavenging biologically important reactive free radicals or other reactive oxygen species (»0 2 ⁇ , H 2 0 2 , »0H, HOCl, ferryl, peroxyl, peroxynitrite, and alkoxyl) , or by preventing their formation, or by catalytically converting the free radical or other reactive oxygen species to a less reactive species.
  • An antioxidant salen-transition metal complex of the invention generally has detectable SOD activity.
  • a salen-transition metal complex of the invention has antioxidant activity if the complex, when added to a cell culture or assay reaction, produces a detectable decrease in the amount of a free radical, such as superoxide, or a nonradical reactive oxygen species, such as hydrogen peroxide, as compared to a parallel cell culture or assay reaction that is not treated with the complex.
  • Suitable concentrations i.e., efficacious dose
  • Antioxidant salen metal complexes of the invention may have glutathione peroxidase activity.
  • a "salen-transition metal complex” refers to a compound having a structure according to Structure I, Structure II, Structure III, or Structure IV, Structure V, Structure VI, Structure VII, Structure VIII, Structure IX (see, infra) or any of the structures Cl, C4, C6, C7, C9, CIO, Cll, C12, C15, C17, C20, C22, C23, C25, C27, C28, C29, and C30 as shown in Fig. 3 and infra , preferably having a structure corresponding to one of the structures shown in Fig.
  • free radical-associated disease refers to a pathological condition of an individual that results at least in part from the production of or exposure to free radicals, particularly oxyradicals, and other reactive oxygen species in vivo.
  • free radical associated disease encompasses pathological states that are recognized in the art as being conditions wherein damage from free radicals or reactive oxygen species is believed to contribute to the pathology of the disease state, or wherein administration of a free radical inhibitor (e.g., desferrioxamine) , scavenger (e.g., tocopherol, glutathione), or catalyst (e.g., SOD, catalase) is shown to produce a detectable benefit by decreasing symptoms, increasing survival, or providing other detectable clinical benefits in treating or preventing the pathological state.
  • a free radical inhibitor e.g., desferrioxamine
  • scavenger e.g., tocopherol, glutathione
  • catalyst e.g., SOD, catalase
  • the disease states discussed herein are considered free radical-associated diseases (e.g., ischemic reperfusion injury, inflammatory diseases, systemic lupus erythematosis, myocardial infarction, stroke, traumatic hemorrhage, spinal cord trauma, Crohn's disease, autoimmune diseases (e.g., rheumatoid arthritis, diabetes), cataract formation, uveitis, emphysema, gastric ulcers, oxygen toxicity, neoplasia, undesired cell apoptosis, radiation sickness, and other pathological states discussed in the Background section and infra) .
  • free radical-associated diseases e.g., ischemic reperfusion injury, inflammatory diseases, systemic lupus erythematosis, myocardial infarction, stroke, traumatic hemorrhage, spinal cord trauma, Crohn's disease, autoimmune diseases (e.g., rheumatoid arthritis, diabetes), cataract formation,
  • SOD mimetic As used herein the terms “SOD mimetic”, “SOD mimic”, “superoxide dismutase mimetic”, and “superoxide catalyst” refer to compounds which have detectable catalytic activity for the dismutation of superoxide as determined by assay.
  • an SOD mimetic possesses at least about 0.001 percent of the SOD activity of human Mn-SOD or Zn,Cu-SOD, on a molar basis, as determined by standard assay methods and/or has at least 0.01 unit of SOD activity per mM according to the SOD assay used hereinbelow, preferably at least 1 unit of SOD activity per mM.
  • alkyl refers to a cyclic, branched, or straight chain alkyl group containing only carbon and hydrogen, and unless otherwise mentioned, contain one to twelve carbon atoms. This term is further exemplified by groups such as methyl, ethyl, n-propyl, isobutyl, t-butyl, pentyl, pivalyl, heptyl, adamantyl, and cyclopentyl.
  • Alkyl groups can either be unsubstituted or substituted with one or more substituents, e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.
  • substituents e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.
  • lower alkyl refers to a cyclic, branched or straight chain monovalent alkyl radical of one to six carbon atoms. This term is further exemplified by such radicals as methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, i-butyl (or 2-methylpropyl) , cyclopropylmethyl, i-amyl, n-amyl, and hexyl.
  • aryl refers to a monovalent unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) , which can optionally be unsubstituted or substituted with, e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.
  • substituted alkoxy refers to a group having the structure -O-R, where R is alkyl which is substituted with a non-interfering substituent.
  • arylalkoxy refers to a group having the structure -O-R-Ar, where R is alkyl and Ar is an aromatic substituent.
  • Arylalkoxys are a subset of substituted alkoxys. Examples of preferred substituted alkoxy groups are: benzyloxy, napthyloxy, and chlorobenzyloxy.
  • aryloxy refers to a group having the structure -O-Ar, where Ar is an aromatic group.
  • a preferred aryloxy group is phenoxy.
  • heterocycle refers to a monovalent saturated, unsaturated, or aromatic carbocyclic group having a single ring (e.g.
  • morpholino pyridyl or furyl
  • multiple condensed rings e.g., indolizinyl or benzo[b]thienyl
  • having at least one heteroatom defined as N, O, P, or S, within the ring, which can optionally be unsubstituted or substituted with, e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.
  • Heteroaryl or “HetAr” refers to an aromatic heterocycle.
  • Arylalkyl refers to the groups -R-Ar and -R-HetAr, where Ar is an aryl group, HetAr is a heteroaryl group, and R is straight-chain or branched-chain aliphatic group. Examples of arylalkyl groups include benzyl and furfuryl.
  • Arylalkyl groups can optionally be unsubstituted or substituted with, e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.
  • halo or halide refers to fluoro, bromo, chloro and iodo substituents.
  • OBn means benzyloxy.
  • amino refers to a chemical functionality -NR'R", where R' and R" are independently hydrogen, alkyl, or aryl.
  • quaternary amine refers to the positively charged group -N + R'R"R" 1 , where R' , R", and R'" are independently selected and are alkyl or aryl.
  • a preferred amino group is -NH 2 .
  • silyl refers to organometallic substituents, wherein at least one silicon atom is linked to at least one carbon atom; an example of a silyl substituent is the tri ethylsilyl substituent, (CH 3 ) 3 Si-.
  • pharmaceutical agent or drug refers to a chemical compound or composition capable of inducing a desired therapeutic effect when properly administered to a patient.
  • the catalytic activity of salen-metal complexes to interconvert epoxides may also be used to advantage to scavenge or prevent formation in vivo of cytotoxic and/or carcinogenic epoxide species, such as may be formed by the cytochrome P-450 monooxygenation system (e.g., benzo-[a]- pyrene diol epoxide) .
  • Catalytic salen-metal complexes may be advantageously included into foodstuffs or dietary supplements (or administered in other forms) to individuals who are at risk of exposure to polycyclic hydrocarbon chemical carcinogens, such as workers in the petrochemical industry and dyestuff manufacture.
  • catalytically active salen- metal complexes may be formulated for administration to smokers (including passive smokers) to enhance detoxification of reactive epoxides formed from cigarette smoke.
  • the antioxidant salen metal complexes of the invention can find use to partially or totally arrest the progression of neurodegenerative diseases. For example, mutations in Cu/Zn superoxide dismutase have been reported to be strongly associated with amyotrophic lateral sclerosis
  • ALS ALS
  • Antioxidant salen metal complexes of the present invention can be used for treatment and prophylaxis of such neurodegenerative diseases (e.g. , ALS, MS) .
  • the salen-transition metal complex has the following structure:
  • M is a transition metal ion, preferably Mn; A is an anion, typically Cl; and n is either 0, 1, or 2.
  • X ⁇ X 2 , X 3 and X 4 are independently selected from the group consisting of hydrogen, silyls, aryls, arylalkyls, primary alkyls, secondary alkyls, tertiary alkyls, alkoxys, aryloxys, aminos, quaternary amines, heteroatoms, and hydrogen; typically X 2 and X 3 are from the same functional group, usually hydrogen, quaternary amine, or tertiary butyl, and X 2 and X 4 are typically hydrogen.
  • Y-_, Y 2 , Y 3 , Y 4 , Y 5 , and Y 5 are independently selected from the group consisting of hydrogen, halides, alkyls, aryls, arylalkyls, silyl groups, aminos, alkyls or aryls bearing heteroatoms; aryloxys, alkoxys, and halide; preferably, Y x and Y 4 are alkoxy, halide, or amino groups. Typically, Y ⁇ and Y 4 are the same.
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, CH 3 , C 2 H 5 , C 6 H 5 , O-benzyl, primary alkyls, fatty acid esters, substituted alkoxyaryls, heteroatom-bearing aromatic groups, arylalkyls, secondary alkyls, and tertiary alkyls.
  • At least one of the X x and X 3 sites, and preferably both X 1 and X 3 include a substituent selected from the group of blocking substituents consisting of secondary or tertiary alkyl groups, aryl groups, silyl groups, heterocycles, and alkyl groups bearing heteroatom substituents such as alkoxy or halide.
  • the X- ⁇ and X 3 sites bear the same substituent, which substituent is most preferably a tertiary alkyl group, such as tertiary butyl.
  • X 2 and X 4 are selected from a group of non-blocking substituents such as H, CH 3 , C 2 H 5 , and primary alkyls, most preferably, H.
  • X ⁇ , X 2 , X 3 , and X 4 can be selected from the group of blocking substituents.
  • R 1# R 2 , R 3 and R 4 are selected from a group consisting of H, CH 3 , C 2 H 5 , and primary alkyls.
  • this group will be referred to as the non-blocking group.
  • R- ⁇ is selected from the non-blocking group
  • R 2 and R 3 are preferably selected from the blocking group, and typically R 2 and R 3 are identical and are phenyl or benzyloxy.
  • R 2 is selected from the non- blocking group
  • R 2 and R 4 are preferably selected from the blocking group.
  • R 3 is selected from the non- blocking group
  • R 2 and R 4 are preferably selected from the blocking group.
  • R 4 is selected from the non- blocking group
  • R 2 and R 3 are preferably selected from the blocking group.
  • Phenyl and benzyloxy are particularly preferred blocking groups for substitution at any of R 1 , R 2 , R 3 and R 4 .
  • the blocking groups selected are identical.
  • a preferred class of embodiments have R ⁇ and R 4 as benzyloxy or phenyl and R 2 and R 3 as hydrogen.
  • one class of embodiments of the first aspect of the invention requires that, of the four sites available for substitution on the two carbon atoms adjacent to nitrogen, at least one or two of these preferably will include a substituent from the non-blocking group.
  • the non-blocking substituent is either hydrogen or methyl, but most preferably, hydrogen.
  • the blocking substituent is either a phenyl group, a benzyloxy, or a tertiary butyl group, more preferably a phenyl group or a benzyloxy group, most usually a phenyl group.
  • Y 3 and Y 6 are hydrogen, methyl, alkyl, or aryl. More preferably, they are hydrogen or methyl. Most preferably, they are hydrogen.
  • the Y l t Y 2 , Y 4 , and Y 5 sites are selected independently and are preferably occupied by hydrogen, although these sites may also he occupied by substituents independently selected from the group consisting of hydrogen, halides, alkyls, aryls, alkoxy groups, substituted alkoxy groups, nitro groups, and amino groups.
  • ⁇ and Y 4 are preferably occupied by methoxy, ethoxy, chloro, bromo, iodo, primary alkyl, tertiary butyl, primary amine, secondary amine, or tertiary amine substituents, most preferably methoxy, chloro, tertiary butyl, or methyl.
  • the salen-transition metal complex has the structure: Structure II
  • M is a transition metal ion, preferably Mn, and A is an anion, typically Cl; where at least one of X or X 2 is selected from the group consisting of aryls, primary alkyls, secondary alkyls, tertiary alkyls, and heteroatoms; where at least one of X ⁇ or X 3 is selected from the group consisting of aryls, primary alkyls, secondary alkyls, tertiary alkyls, arylalkyls, heteroatoms, and hydrogen, preferably tertiary butyl or hydrogen; and where Y l r Y 2 , Y 3 , Y 4 • 5' • 6' J l ' J 2 ' J 3'
  • J 4' J 5 ' '6 ' Z 7 , Z 8 , Z 9 , Z 10 , Z 11# and Z 12 are independently selected from the group consisting of hydrogen, halides, alkyls, aryls, amines, alkoxy, substituted alkoxy, arylalkyls, aryloxys, and alkyl groups bearing heteroatoms.
  • Y ⁇ and Y 4 are selected from the group consisting of lower alkyls, alkoxy, halide, and amino groups, more preferably from the group consisting of methoxy, chloro, and primary amine.
  • Y x and Y 4 are methoxy
  • X and X 3 are independently selected and are hydrogen or tertiary butyl, and the remaining substituents are hydrogen.
  • the salen-transition metal has the following structure:
  • M is a transition metal ion, typically Mn, and A is an anion, typically Cl; where n is either 4, 5, or 6; where X l f X 2 , X 3 , and X 4 are independently selected from the group consisting of aryls, arylalkyls, aryloxys, primary alkyls, secondary alkyls, tertiary alkyls, alkoxy, substituted alkoxy, heteroatoms, aminos, quaternary amines, and hydrogen; preferably, at least one of ⁇ or X 3 are selected from the group consisting of aryls, primary alkyls, secondary alkyls, tertiary alkyls, quaternary amines, arylalkyls, heteroatoms, and hydrogen; preferably X-, ⁇ and X 3 are identical and are hydrogen or tertiary butyl; where Y , Y 2 , Y 3 , Y 4 , Y 5
  • R 2 and R 4 are independently selected from the group consisting of hydrogen, halides, primary alkyls, secondary alkyls, tertiary alkyls, fatty acid esters, alkoxys, or aryls.
  • R 2 and R 4 are identical; more preferably R 2 and R 4 are hydrogen.
  • Y ⁇ and Y 2 are independently selected from the group consisting of methoxy, ethoxy, methyl, ethyl, t-butyl, chloro, bromo, iodo, amino, quaternary amine, alkylamino, dialkylamino, and hydrogen; R ⁇ and R 2 are independently selected from the group consisting of: phenyl, benzyloxy, chlorobenzyloxy, hydrogen, amino, quaternary amune, or fatty acid ester.
  • Y 2 and Y 2 are identical.
  • R 1 and R 2 are selected independently from the group consisting of: phenyl, benzyloxy, chlorobenzyloxy, hydrogen, amino, quaternary amine, or fatty acid ester.
  • R 1 and R 2 are identical, Structure VI
  • Y 2 and Y 2 are independently selected from the group consisting of methoxy, ethoxy, methyl, ethyl, t-butyl, chloro, bromo, iodo, amino, quaternary amine, alkylamino, dialkylamino, and hydrogen;
  • R ⁇ and R 2 are selected independently from the group consisting of: phenyl, benzyloxy, chlorobenzyloxy, hydrogen, amino, quaternary amine, or fatty acid ester.
  • Y and Y 2 are identical, and R ⁇ and R 2 are identical.
  • X is selected from the group consisting of methoxy, ethoxy, methyl, ethyl, t-butyl, chloro, bromo, iodo, amino, quaternary amine, alkylamino, dialkylamino, and hydrogen;
  • Y is selected from the group consisting of t-butyl, quaternary amine, amino, and hydrogen.
  • R 2 are independently selected from the group consisting of aryloxys, alkoxys, aryls, and hydrogen; R' and R" are independently selected from the group consisting of alkyls, aryls, and hydrogen.
  • at least one of the amino groups is protonated at physiological pH (i.e., pH 7.3- 7.8).
  • Preferred R* or R" alkyls include but are not limited to: methyl, ethyl, and propyl.
  • Preferred R ⁇ and R 2 aryloxys include but are not limited to benzyloxy and chlorobenzyloxy.
  • Preferred R ⁇ and R 2 alkoxys include but are not limited to ethoxy and methoxy.
  • a preferred subgenus of Structure VIII includes, but is not limited to:
  • R is selected from the group consisting of alkyls and hydrogen.
  • at least one of the amino groups are protonated at physiological pH (i.e., pH 7.3-7.8).
  • antioxidant salen-transition metal complexes for formulation in pharmaceutical compositions, dietary supplements, foodstuff preservatives, cosmetics, sunburn preventatives, and other compositions of the invention, and are referenced by structure number (e.g., Cl through C30) for clarity throughout.
  • compositions of the present invention comprise a therapeutically or prophylactically effective dose of at least one salen derivative-based complex of a transition metal ion.
  • the term "salen” is used herein to refer to those ligands typically formed through a condensation reaction of two molecules of a salicylaldehyde derivative with one molecule of a diamine derivative. While salen ligands are formed from ethylenediamine derivatives, propyl and butyl diamines may also be used to give analogous salen and salen derivatives. Salen derivatives are preferred and their general structure is shown in Fig. 1. A salen derivative where n is 0 is shown in Fig. 2.
  • the two nitrogens and the two oxygens are oriented toward the center of the salen ligand and thus provide a complexing site for the transition metal ion M.
  • this metal ion is selected from the group consisting of Mn, Cr, Fe, Ni, Co, Ti, V, Ru, and Os. More preferably, the transition metal ion is selected from the group consisting of Mn, Mg, Cr, Fe, Ni, and Co. Most preferably, the metal ion is Mn.
  • the anion is selected from the group consisting of PF 6 , (aryl) 4 , BF 4 , B(aryl) 4 , halide, acetate, triflate, tosylate, with halide or PF 6 being more preferred. and chloride being most preferred.
  • Fig. 1 also shows the many sites available for substitution on the salen ligand. Of these sites, it is believed that R l f R 2 , R 3 , R , and X l r X 2 , X 3 , X 4 , Y 3 and Y 6 are the most important in this first salen-transition metal complex.
  • Structures I, III, IV, VI, VII, and VIII may have independently selected fatty acid ester substituents at the R l f R 2 , R 3 , and R 4 positions.
  • the fatty acid esters typically occupy no more than two substituent positions and are usually identical.
  • the unsaturated acids occur in isomeric forms due to the presence of the one or more unsaturated positions.
  • the compounds of the present invention are intended to include the individual double bond isomers, as well as mixtures thereof.
  • the fatty acid esters of the present invention can be obtained by known acylation techniques. See, e.g., March, Advanced Organic Chemistry, 3rd Ed., John Wiley & Sons, New York (1985), pp. 299, 348-351, and 353-354, incorporated herein by reference.
  • Figure 3 shows structures of preferred antioxidant salen-transition metal complexes of the invention.
  • Example antioxidant salen-transition metal complexes are shown in Fig. 3.
  • Compounds Cl, C4, C6, C7, C9, CIO, Cll, and C12 are particularly preferred for formulation in pharmaceuticals and other antioxidant compositions of the invention. It is believed that C7 is particularly preferred because of its facile preparation and relatively hydrophilic nature which is well-suited to pharmaceutical usage.
  • a preferred salen-transition metal complex having high superoxide dismutase activity is the C12 compound having the structure: additional preferred congeners of C12 are:
  • a particularly preferred antioxidant salen-metal complex of the invention is C7:
  • Antioxidant salen-transition metal complexes generally have detectable superoxide dismutase activity and preferably also have catalase activity.
  • C7 is both simple to prepare and relatively hydrophilic, properties which make it particularly well-suited for pharmaceutical use and formulation in aqueous solution.
  • the relatively hydrophilic nature of C7 can be used to advantage in providing antioxidant salen-metal complexes that are readily absorbed and transported in the human body.
  • One advantageous pharmacokinetic property of C7 is believed to be the capacity to cross the blood-brain barrier efficiently.
  • the preferred route to prepare the antioxidant salen-transition metal complexes of the present invention is a condensation reaction with the substituted salicylaldehyde and the substituted diamine.
  • quantities of these compounds are reacted in a 2 to 1 molar ration in absolute ethanol.
  • the solutions are refluxed typically for 1 hour, and the salen ligand is either precipitated in analytically pure form by addition of water, or the metal complex is generated directly by addition of the metal as its acetate, halide, or triflate salt.
  • the starting diamine is R,R- or S,S-1,2-diamino- 1,2-diphenylethane and the starting salicylaldehyde is 3-tert- butylsalicylaldehyde.
  • a solution of 2.0 mmol of 3-tert- butylsalicylaldehyde in 3 ml of absolute ethanol is added dropwise to a solution of 1.0 mmol of (R,R) -1,2-diamino-l,2- diphenylethane in 5 ml of ethanol.
  • the reaction mixture is heated to reflux for 1 h and then 1.0 mmol of Mn(Oac) 2 -4H 2 0 is added in one portion to the hot (60°C) solution.
  • the color of the solution immediately turns from yellow to brown upon addition. It is refluxed for an additional 30 min and then cooled to room temperature.
  • a solution of 10% NaCl (5ml) is then added dropwise and the mixture stirred for 0.5h.
  • the solvents are then removed in vacuo and the residue is triturated with 50 ml of CH 2 -C1 2 and 50 ml of H 2 0.
  • the organic layer is separated and the brown solution is washed with saturated NaCl.
  • the SOD activity of the prepared salen-Mn complexes is determined according to standard assay methods for SOD activity known in the art and exemplified infra .
  • Salen-metal complexes having at least 0.01 unit of SOD activity per millimole/liter in aqueous solution are antioxidant salen- metal complexes; preferably antioxidant salen-metal complexes have at least about 1 unit of SOD activity per millimole/liter; and more preferably have at least about 100 units of SOD activity per millimole/liter; frequently having more that 500 to 1000 units of SOD activity per mM or more.
  • the SOD mimetic salen- metal complex also possesses detectable catalase activity (e.g., C4, C7, C9, CIO, Cll, C12); typically at least 10 units of catalase activity per mM, and frequently at least 100 units of catalase activity per mM.
  • detectable catalase activity e.g., C4, C7, C9, CIO, Cll, C12
  • compositions comprising an antioxidant salen-transition metal complex of the present invention are useful for topical and parenteral administration, i.e.. subcutaneously, intramuscularly or intravenously.
  • the finding that salen-metal complexes possess SOD activity in vitro as well as functioning in vivo indicates that antioxidant salen-metal complexes are suitable SOD mimetics for pharmaceutical use.
  • the antioxidant salen-metal complexes are suitable for administration to mammals, including human patients and veterinary patients.
  • compositions for parenteral administration will commonly comprise a solution of an antioxidant salen- transition metal complex or a cocktail thereof dissolved in an acceptable carrier, preferably an aqueous carrier. Since many of the salen-Mn complexes of the invention are lipophilic, it is preferable to include in the carrier a hydrophobic base (e.g., polyethylene glycol, Tween 20).
  • a hydrophobic base e.g., polyethylene glycol, Tween 20.
  • aqueous carriers can be used, e.g. , water, buffered water, 0.4% saline, 0.3% glycine and the like. These solutions are sterile and generally free of particulate matter. These compositions may be sterilized by conventional, well known sterilization techniques.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, etc.
  • concentration of the antioxidant salen-transition metal complex(es) in these formulations can vary widely, i.e. , from less than about 1 nM, usually at least about O.lmM to as much as 100 mM and will be selected primarily based on fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected. Most usually, the antioxidant salen-metal complex is present at a concentration of 0.1 mM to 10 mM.
  • a typical formulation for intravenous injection comprises a sterile solution of an antioxidant salen-metal complex (e.g., C7) at a concentration of 5 mM in Ringer's solution.
  • an antioxidant salen-metal complex e.g., C7
  • a hydrophobic vehicle may be used, or that an aqueous vehicle comprising a detergent or other lipophilic agent (e.g., Tween, NP-40, PEG); alternatively, the antioxidant salen complexes may be administered as a suspension in an aqueous carrier, or as an emulsion.
  • a typical pharmaceutical composition for intramuscular injection could be made up to contain 1 ml sterile buffered water, and about 1-100 mg of antioxidant salen-transition metal comple (es).
  • a typical composition for intravenous infusion can be made up to contain 250 ml of sterile Ringer's solution, and about 100-1000 mg of antioxidant salen-transition metal complex(es) .
  • Lipophilic agents may be included in formulations of lipophilic salen- metal complexes. Actual methods for preparing parenterally administrable compositions will be known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science, 15th Ed. , Mack Publishing Company, Easton, Pennsylvania (1980) , which is incorporated herein by reference.
  • a typical pharmaceutical composition for topical application can be made with suitable dermal ointments, creams, lotions, ophthalmic ointments and solutions, respiratory aerosols, and other excipients.
  • Excipients should be chemically compatible with the antioxidant salen-transition metal complex(es) that are the active ingredient(s) of the preparation, and generally should not increase decomposition, denaturation, or aggregation of active ingredient(s) .
  • excipients will have lipophilic components such as oils and lipid emulsions.
  • the antioxidant salen-transition metal complex(es) of this invention can be lyophilized for storage and reconstituted in a suitable carrier prior to use. It will be appreciated by those skilled in the art that lyophilization and reconstitution can lead to varying degrees of antioxidant activity loss, and that use levels may have to be adjusted to compensate.
  • compositions containing the present antioxidant salen-transition metal complex(es) or cocktails thereof can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already affected by the particular free radical- associated disease, in an amount sufficient to cure or at least partially arrest the condition and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective dose” or "efficacious dose.”
  • Amounts effective for this use will depend upon the severity of the condition, the general state of the patient, and the route of administration, but generally range from about 1 mg to about lOg of antioxidant salen-transition metal complex(es) per dose, with dosages of from 10 mg to 2000 mg per patient being more commonly used.
  • a antioxidant salen metal complex e.g., C7
  • at least about lOmg to 500 mg of antioxidant salen-metal complex(es) may be administered by intrapericardial injection to provide elevated local concentrations of SOD activity in the myocardium.
  • compositions containing the antioxidant salen-transition metal complex(es) or cocktails thereof are administered to a patient not already in a disease state to enhance the patient's resistance or to retard the progression of disease.
  • Such an amount is defined to be a "prophylactically effective dose.”
  • the precise amounts again depend upon the patient's state of health and general level of immunity, but generally range from 1 mg to 10 g per dose, especially 10 to 1000 mg per patient.
  • a typical formulation of an antioxidant salen-metal complex such as C7 will contain between about 25 and 250 mg of the salen-metal complex in a unit dosage form.
  • compositions can be carried out with dose levels and dosing pattern being selected by the treating physician.
  • the pharmaceutical formulations should provide a quantity of the antioxidant salen-transition metal complex(es) of this invention sufficient to effectively treat the patient.
  • Kits can also be supplied for use with the subject antioxidant salen-transition metal complex(es) for use in the protection against or therapy for a free radical-associated disease.
  • the subject composition of the present invention may be provided, usually in a lyophilized form or aqueous solution in a container, either alone or in conjunction with additional antioxidant salen-transition metal complex(es) of the desired type.
  • the antioxidant salen- transition metal complex(es) are included in the kits with buffers, such as Tris, phosphate, carbonate, etc. , stabilizers, biocides, inert proteins, e.g.. serum albumin, or the like, and a set of instructions for use.
  • buffers such as Tris, phosphate, carbonate, etc.
  • stabilizers such as Tris, phosphate, carbonate, etc.
  • biocides such as Tris, phosphate, carbonate, etc.
  • inert proteins e.g.. serum albumin, or the like
  • a set of instructions for use Generally, these materials will be present in less than about 5% wt. based on the amount of antioxidant salen-transition metal comple (es), and usually present in total amount of at least about 0.001% based again on the concentration.
  • Salen-Mn complexes preferably compound C12 or C7, can be incorporated into a hypothermic cardioplegia solution at a concentration of at least about 1 mM into a solution formulation according to Amano et al. (1982) Jpn. J. Surq. 12: 87, incorporated herein by reference. Most preferably, C7 is included in the cardioplegia solution.
  • the dosage of SOD-mimetic salen-metal complex(es) will vary with each particular application.
  • the composition is administered either systemically or topically.
  • Systemic administration includes per os and parenteral routes; topical administration includes in situ applications.
  • the in situ means includes, for example, administering an SOD-mimetic salen-metal complex by endoscopic bolus wash and/or paravenous injection, or in the case of lower GI treatments, by enema.
  • Parenteral routes may include, for example, subcutaneous, intradermal, intramuscular, and intravenous routes.
  • the amount of SOD-mimetic salen-metal complex(es) will range from about 2 to 5,000 mg or more, typically 10 to 1000 mg, depending on the administration interval and route, which can range from a single oral dose, parenteral dose and/or topical dose to multiple oral doses, parenteral doses, and/or topical doses over a few days or greater than 5 weeks.
  • the dosage may also vary with the severity of the disease.
  • antioxidant salen-transition metal complexes of the invention are employed to modulate the expression of naturally-occurring genes or other polynucleotide sequences under the transcriptional control of an oxidative stress response element (e.g., an antioxidant responsive element, ARE) , such as an antioxidant response element of a glutathione S-transferase gene or a NAD(P)H:quinone reductase gene (Rozen et al. (1992) Arch. Biochem. Biophys. 292: 589; Favreau and Pickett (1991) J. Biol. Chem. 266: 4556; Rushmore and Pickett (1991) Methods Enzymol.
  • an oxidative stress response element e.g., an antioxidant responsive element, ARE
  • an antioxidant response element of a glutathione S-transferase gene or a NAD(P)H:quinone reductase gene Rosenzymol.
  • Transgenes, homologous recombination constructs, and episomal expression systems comprising a polynucleotide sequence under the transcriptional control of one or more ARE linked to a promoter will be made by those of skill in the art according to methods and guidance available in the art, as will transformed cells and transgenic nonhuman animals harboring such polynucleotide constructs.
  • the antioxidant salen-metal complexes may be used to modulate the transcription of ARE- regulated polynucleotide sequences in cell cultures (e.g., ES cells) and in intact animals, particularly in transgenic animals wherein a transgene comprises one or more AREs as transcriptional regulatory sequences.
  • a dose-response curve is generated by titrating transcription rate of the ARE-controlled polynucleotide sequence against increasing concentrations of antioxidant salen-metal complex(es) , which will reduce the transcription rate induced by oxidant agents (e.g., benzoyl peroxide, glutathione-depleting agent) or oxidative stress.
  • oxidant agents e.g., benzoyl peroxide, glutathione-depleting agent
  • a therapeutically or pharmaceutically effective amount of an antioxidant salen- transition metal complex is administered to a patient to treat or prevent a free radical-associated disease.
  • the required dosage will depend upon the nature of the free radical- associated disease, the severity and course of the disease, previous therapy, the patient's health status and response to the antioxidant salen-transition metal complex, and the judgment of the treating physician.
  • At least one species of antioxidant salen-Mn complex is administered as the sole active ingredient, or in combination with one or more other active ingredients, typically selected from the group consisting of: N-2-mercaptopropionylglycine, N-acetylcysteine, glutathione, dimethyl thiourea, desferrioxamine, mannitol, ⁇ - tocopherol, ascorbate, allopurinol, 21-aminosteroids, calpain inhibitors, glutamate receptor antagonists, tissue plasminogen activator, streptokinase, urokinase, nonsteroidal anti- inflammatory agent, cortisone, and carotenoids.
  • active ingredients typically selected from the group consisting of: N-2-mercaptopropionylglycine, N-acetylcysteine, glutathione, dimethyl thiourea, desferrioxamine, mannitol, ⁇ - tocopherol, ascorbate,
  • Antioxidant salen-Mn complexes may also be administered in conjunction with polypep ides having SOD and/or catalase activity, particularly in view of the capacity of the salen-Mn complexes, unlike SOD polypeptides, to cross the blood-brain barrier and thereby complement systemic SOD administration.
  • the present invention includes a method of treating patients, such as humans, who have a free radical-associated disease with a prophylactically effective or therapeutically effective amount of a antioxidant salen-transition metal complex, typically a salen-Mn complex, preferably C7. This method can be used to treat patients at various stages of their diseases or to prevent development of free radical- associated diseases in patients.
  • the treatment can be administered to prevent or reduce, as a prophylactic, the age-adjusted probability of developing a neoplasm and/or the age-adjusted mortality rate and/or the rate of senescence.
  • the antioxidant salen-metal complexes of the invention can also be administered to patients who are infected with a human immunodeficiency virus (e.g., HIV-l) or who are at risk of becoming infected with a human immunodeficiency virus.
  • the antioxidant salen-metal complexes can prevent or inhibit the induction of HIV-l replication in CD4 + lymphocytes by tumor necrosis factor (TNF) and/or prevent damage to or death of CD4 + cells as a consequence of HIV-l infection.
  • TNF tumor necrosis factor
  • administration of an antioxidant salen-metal complex, such as C7 can inhibit and/or slow the development of HIV-l related pathology and/or can reduce the rate of decline of the CD4 + lymphocyte population in HIV-infected individuals.
  • the antioxidant salen-metal complexes can also inhibit pathology resulting from excessive or inappropriate levels of TNF, both in AIDS and in other conditions (e.g., septic shock) .
  • a dosage of about 50 to 5000 mg will be administered to a patient with HIV and/or with excessive or inappropriate levels of TNF, either in single or multiple doses, to reduce or retard the development of pathology and clinical symptoms.
  • Antioxidant salen-metal complexes may be administered therapeutically to treat viral diseases other than HIV.
  • antioxidant salen-transition metal complexes Since oxidative damage occurs proportionately to the abundance of free radicals and reactive oxygen species, it is expected that administration of antioxidant salen-transition metal complexes at even low levels will confer a protective effect against oxidative damage; thus it is expected that there is no threshold level below which antioxidant salen-Mn complexes are ineffective.
  • a suitable effective dose of the antioxidant salen- Mn complex will be in the range of 0.01 to 1000 milligram (mg) per kilogram (kg) of body weight of recipient per day, preferably in the range of 1 to 100 mg per kg of body weight per day.
  • the desired dosage is preferably presented in one, two, three, four or more subdoses administered at appropriate intervals throughout the day. These subdoses can be administered as unit dosage forms, for example, containing 5 to 10,000 mg, preferably 10 to 1000 mg of active ingredient per unit dosage form.
  • the composition used in these therapies can be in a variety of forms.
  • compositions also preferably include conventional pharmaceutically acceptable carriers and adjuvants which are known to those of skill in the art. See, e.g., Remington's Pharmaceutical Sciences. Mack Publishing Co.: Easton, PA, 17th Ed. (1985).
  • administration will be by oral or parenteral (including subcutaneous, intramuscular, intravenous, and intradermal) routes, or by topical application or infusion into a body cavity, or as a bathing solution for tissues during surgery.
  • the methods of this invention can be used in combination with other antioxidant agents that have SOD activity, catalase activity, GSH-Px activity, or are free radical scavengers or inhibitors of free radical formation. While it is possible to administer the active ingredient of this invention alone, it is believed preferable to present it as part of a pharmaceutical formulation.
  • the formulations of the present invention comprise at least one compound of this invention in a therapeutically or pharmaceutically effective dose together with one or more pharmaceutically or therapeutically acceptable carriers and optionally other therapeutic ingredients.
  • Various considerations are described, e.g., in Gilman et al. (eds) (1990) Goodman and Gilman's: The
  • compositions will be administered by parenteral or oral administration for prophylactic and/or therapeutic treatment.
  • the pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration.
  • unit dosage forms suitable for oral administration include powder, tablets, pills, capsules, and dragees.
  • the pharmaceutical compositions will often be administered intravenously.
  • this invention provides compositions for intravenous administration which comprise a solution of the compound dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • an aqueous carrier e.g., water, buffered water, 0.4% saline, and the like.
  • the antioxidant salen-metal complex(es) such as C7 or C12, may be dissolved in an organic solvent (e.g., dimethylsulfoxide) and either applied directly or diluted into an aqueous solvent.
  • an organic solvent e.g., dimethylsulfoxide
  • antioxidant salen-metal complexes that are relatively lipophilic e.g.,
  • compositions are dissolved in an organic solvent such as DMSO and, if desired, subsequently diluted into a more polar solvent, such as water.
  • organic solvent such as DMSO
  • a more polar solvent such as water.
  • These compositions will sometimes be sterilized by conventional, well known sterilization techniques, or can preferably be sterile filtered.
  • the resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • compositions can contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, and the like.
  • auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, and the like.
  • nontoxic solid carriers can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 0.001-95% of active ingredient, preferably about 20%.
  • compositions containing the compounds can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already suffering from a disease, as described above, in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as
  • terapéuticaally effective amount or dose Amounts effective for this use will depend on the severity of the disease and the weight and general state of the patient.
  • compositions containing the compounds of the invention are administered to a patient susceptible to or otherwise at risk of a particular disease. Such an amount is defined to be a "prophylactically effective amount or dose.” In this use, the precise amounts again depend on the patient's state of health and weight.
  • conventional non-toxic solid excipients include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, celluloses, glucose, sucrose, magnesium carbonate, and the like may be used.
  • the active compound as defined above may be formulated as suppositories using, for example, triglycerides, for example, the Witepsols, as the carrier.
  • Liquid pharmaceutically administerable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension.
  • a excipient such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, etc.
  • composition or formulation to be administered will, in any event, contain an effective amount of the active compound(s).
  • a pharmaceutically acceptable non-toxic composition is formed by the incorporation of any of the normally employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, celluloses, glucose, sucrose, magnesium, carbonate, and the like.
  • excipients such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, celluloses, glucose, sucrose, magnesium, carbonate, and the like.
  • Such compositions take the form of solutions, suspensions, tablets, capsules, powders, sustained release formulations and the like.
  • Such compositions may contain 0.01-95% active ingredient, preferably 1-70%.
  • Parenteral administration is generally characterized by injection, either subcutaneously, intramuscularly or intravenously.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like.
  • the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, etc.
  • a more recently devised approach for parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. See, e.g., U.S. Patent No. 3,710,795, which is incorporated herein by reference.
  • Antioxidant salen-metal complexes may be administered by transdermal patch (e.g., iontophoretic transfer) for local or systemic application. Once detectable improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved condition is retained. When the symptoms have been alleviated to the desired level, treatment can cease.
  • Antioxidant salen-metal complex(es) can also be added to extravasated blood for transfusion to inhibit oxyradical damage to the blood cells and components during storage; similarly, antioxidant salen-metal complexes can also reduce oxyradical damage to blood cells m vivo.
  • Antioxidant salen-metal complex(es) can also be added to rinse or storage solutions for organs and tissues, such as for organ transplantation or for surgical rinses. For example, excised organs are often placed in a preservation solution prior to transplant into a recipient.
  • At least one species of antioxidant salen-metal complex in a preservation solution is desirable for reducing damage due to ischemia during storage and reperfusion injury following reimplantation in the recipient.
  • a preservation solution usually at a concentration of about 0.01 mM to 10 mM.
  • Various solutions described in the art are suitable for the inclusion of a salen-metal complex, including but not limited to those described in U.S. Patent 5,145,771; Beyersdorf (1990) Chem Abst. 113: 84849w; U.S. Patent 4,879,283; U.S. Patent 4,873,230; and U.S. Patent 4,798,824, incorporated herein by reference.
  • the antioxidant salen-metal complex is present in the rinse or storage solution at a concentration of about lO ⁇ M to about 10 mM, and most usually is present at 1 mM.
  • a suitable rinse solution comprises Ringer's solution (102 mM NaCl, 4 mM KC1, 3 mM CaCl 2 , 28 mM sodium lactate, pH 7.0) or Ringer's solution with 0.1 mM adenosine, and the antioxidant salen-Mn complex C7 at a final concentration of 1 mM.
  • the rinse solution can further comprise additional antioxidants (e.g., glutathione, allopurinol) .
  • Preservation or rinse solutions containing an antioxidant salen-metal complex can be used to provide enhanced storage or irrigation of organs (e.g., kidney, liver, pancreas, lung, fetal neural tissue, heart, vascular grafts, bone, ligament, tendon, skin) which is believed to enhance the viability of the tissue and increase resistance to oxidative damage (e.g., as a consequence of ischemia/reperfusion) .
  • organs e.g., kidney, liver, pancreas, lung, fetal neural tissue, heart, vascular grafts, bone, ligament, tendon, skin
  • a catalytically active salen-metal complex may be used to advantage in generating free radicals, such as superoxide, in local areas (e.g., for acne treatment, skin cancer treatment, papillomas) or in cell cultures or transgenic animals harboring a transgene under the transcriptional control of a ARE.
  • free radicals e.g., superoxide
  • the capacity of the antioxidant salen-metal complexes to catalyze the decomposition of reactive oxygen species can be used to advantage to inhibit or slow damage to biological tissues and cells.
  • benzoyl peroxide is a widely used treatment for acne lesions; excessive or inappropriate application of benzoyl peroxide (e.g., accidental application to the eyes) may be treated by local (or if desired, systemic) administration of an antioxidant salen-metal complex (e.g., C7) .
  • oxyradical-induced damage to connective tissues attendant to exposure to UV light, cigarette smoking, and senescence may be reduced by administration of an antioxidant salen-metal complex approximately concomitant with the exposure to UV light, cigarette smoking, or other oxyradical-generating process (e.g., cellular senescence).
  • Antioxidant salen-transition metal complexes typically antioxidant salen-Mn complexes, such as compound C7, are used to protect cells and tissues from free radical- producing agents, such as ionizing radiation and chemotherapeutic agents (e.g., bleomycin) .
  • free radical- producing agents such as ionizing radiation and chemotherapeutic agents (e.g., bleomycin) .
  • a protective dosage comprising at least about l ⁇ g of salen-Mn complex/kg bodyweight is administered by one or more of several routes (e.g., oral, intraveneous, intraperitoneal, intragastric lavage, enema, portal vein infusion, topical, or inhalation of mist) , preferably by injection of liposomes or immunoliposomes for targeted delivery of the antioxidant salen-Mn complexes to protect normal cells, for example, against free radical toxicity associated with chemotherapy or radiotherapy of a neoplasm.
  • routes e.g., oral, intraveneous, intraperitoneal, intragastric lavage, enema, portal vein infusion, topical, or inhalation of mist
  • liposomes or immunoliposomes for targeted delivery of the antioxidant salen-Mn complexes to protect normal cells, for example, against free radical toxicity associated with chemotherapy or radiotherapy of a neoplasm.
  • the antioxidant salen-transition metal complexes are preferably preadministered to the patient prior to the commencement of the chemotherapy and/ or radiotherapy, usually within about 24 hours of commencement, and preferably within about 3-6 hours of commencement of the chemotherapy and/ or radiotherapy.
  • Antioxidant salen-Mn may be continually administered to the patient during the course of therapy.
  • a solution of an antioxidant salen- metal complex can be encapsulated in micelles to form immunoliposomes (U.S. Patent 5,043,164, U.S. Patent 4,957,735, U.S. Patent 4,925,661; Connor and Huang (1985) J. Cell Biol. 101: 582; Lasic DD (1992) Nature 355: 279; Novel Drug Delivery (eds. Prescott LF and Nimmo WS: Wiley, New York, 1989) ; Reddy et al. (1992) J. Immunol. 148: 1585; incorporated herein by reference) .
  • the immunoliposomes containing the antioxidant salen-metal species will comprise a targeting moiety (e.g.
  • monoclonal antibody that targets the immunoliposomes to non- neoplastic cells that are otherwise sensitive to radiotherapy or chemotherapy.
  • immunoliposomes having a monoclonal antibody that binds specifically to a hematopoietic stem cell antigen not present on the cancer cells of the individual may be used to target antioxidant salen-metal complexes to hematopoietic stem cells and thereby protect said stem cells against radiotherapy or chemotherapy used to treat the cancer.
  • the chemotherapeutic agent forms free radicals jln vivo (e.g., bleomycin) .
  • Antioxidant salen-Mn complexes are also administered to individuals to prevent radiation injury or chemical injury by free radical generating agents.
  • Military personnel and persons working in the nuclear, nuclear medicine, and/or chemical industries may be administered salen-Mn complexes prophylactically.
  • Antioxidant salen-metal complexes may also be used as chemoprotective agents to prevent chemical carcinogenesis; particularly by carcinogens which form reactive epoxide intermediates (e.g., benzo-[a]-pyrene, benzanthracene) and by carcinogens or promoting agents which form free radicals directly or indirectly (e.g., phenobarbital, TPA, benzoyl peroxide, peroxisome proliferators: ciprofibrate, clofibrate) . Persons exposed to such chemical carcinogens are pretreated with an antioxidant salen-metal complex to reduce the incidence or risk of developing neoplasia.
  • Antioxidant salen-metal complexes can also be formulated into a lipophilic base (or, if desired, an aqueous carrier) for topical application in cosmetics or sunburn- prevention creams and lotions.
  • a typical cosmetic or sunburn- prevention cream or lotion will comprise about between 1 mg to 50 mg of antioxidant salen-metal complex per gram of cosmetic or sunburn-prevention cream or lotion.
  • Antioxidant salen-metal complexes may also be administered to deep-divers or individuals exposed to hyberbaric environments were oxygen toxicity presents a health risk. Administration of an efficacious dose of an antioxidant salen-metal complex to an individual may permit the breathing or hyberbaric and/or oxygen-enriched gases with a reduced risk of oxygen toxicity.
  • an efficacious dosage of an antioxidant salen-metal complex can reduced toxicity and biological damage associated with exposure to ozone.
  • Prophylactic administration of an antioxidant salen-metal complex to humans who are or will be exposed to ozone is expected to confer an enhanced resistance to ozone toxicity, such as the ozone-induced lung damage noted in geographical areas with high ozone levels (e.g. , Los Angeles) .
  • antioxidant salen- transition metal complexes preferably salen-Mn complexes
  • the compounds of the invention inhibit the deleterious effects of ischaemia (coronary infarction and reperfusion in the heart; transient myocardial or CNS ischemia during surgery) without direct depressant effects on myocardial contractility.
  • the compounds are effective in animal models for cardiovascular and CNS diseases, and will be useful for the treatment of myocardial infarction, stroke, brain injury, and transplant surgery, particularly with reperfusion of infarcted areas, arrhythmias, variant and exercise-induced angina, congestive heart failure, stroke and other circulatory disorders, in mammals, particularly in human beings.
  • the salen-Mn complexes are also included in preservation solutions used to bathe excised organs (e.g. , heart, kidney, pancreas, liver, lung) during transport and storage of the excised organ prior to transplantion surgery, including skin grafting and corneal grafting.
  • the preservation solutions will typically comprise at least about 1 ⁇ M of an antioxidant salen-metal complex, preferably at least about 1 mM of an antioxidant salen-metal complex.
  • Administration of the active compound and salts described herein can be via any of the accepted modes of administration for therapeutic agents. These methods include oral, parenteral, transdermal, subcutaneous and other systemic modes.
  • the preferred method of administration is oral, except in those cases where the subject is unable to ingest, by himself, any medication. In those instances it may be necessary to administer the composition parenterally.
  • composition comprises an antioxidant salen-metal species having an amino substituent that can be protonated at physiological pH
  • antioxidant salen-metal complex is dissolved or suspended in a solution having a pH at which the amino substituent is protonated.
  • the amount of active compound administered will, of course, be dependent on the subject being treated, the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. However, an effective dosage is in the range of 0.01-50 mg/kg/day, preferably 0.5-25 mg/kg/day. For an average 70 kg human, this would amount to 0.7-3500 mg per day, or preferably about 35-1750 mg/day.
  • the SOD activity of the compounds was determined by evaluating the inhibition of the reduction of cytochrome C produced by the oxygen free radical generating system, xanthine plus xanthine oxidase. Cytochrome C reduction is monitored spectrophotometrically at 550 nm according to the method described in Darr et al. (1987) Arch. Biochem. Biophvs. 258: 351, incorporated herein by reference. The concentration of xanthine oxidase is adjusted such that it produces a rate of reduction of cytochrome C at 550 nm of 0.025 absorbance unit per minute.
  • the amount of SOD activity required to inhibit the rate of cytochrome C reduction by 50 percent is defined as one unit of activity.
  • Salen- metal complexes are identified as antioxidants if they have at least 0.1 unit of activity at a concentration of 1 mM under these standard assay conditions.
  • Catalase activity was measured using a spectrophotometric method in which the decomposition of hydrogen peroxide is monitored at 240 nm according to the method of Aebi et al. (1984) Methods Enzy ol. 105: 121, incorporated herein by reference.
  • One unit of catalase activity is defined as the amount of enzyme (or salen-metal complex) required to decompose 1 ⁇ mole of hydrogen peroxide in one minute.
  • Each of the compounds was formulated in saline and was stable with no loss of activity observed after several weeks of storage at room temperature. Frequently, it is desirable to first dissolve the salen-metal complex in an organic solvent (e.g., DMSO) and then dilute the solution into a more polar solvent such as water. This is particularly preferred for salen-metal species that are relatively hydrophobic (e.g., C12) .
  • Table IV shows the in vitro SOD and catalase activities of the various salen-Mn complexes tested. SOD and catalase activities are expressed as units/mM.
  • a widely used assay to determine the therapeutic potential of molecules in brain ischemia consists of evaluating their ability to prevent irreversible damage induced by an anoxic episode in brain slices maintained under physiological conditions. Rat brain slices were maintained at 35°C in an interface chamber in an artificial cerebrospinal fluid containing: 124 mM NaCl, 3 mM KCl, 1.25 mM KH 2 P0 4 , 3 mM CaCl, 1 mM MgCl 2 , 26 mM NaHC0 3 , 10 mM D-glucose, and 2 mM L- ascorbate, continuously gassed with a mixture of 0 2 :C0 2 (95:5) .
  • the atmosphere of the chamber was also continuously gased with the mixture of 0 2 :C0 2 (95:5), except during the anoxic episode when it was replaced by N 2 .
  • Axons were electrically stimulated and the evoked excitatory post-synaptic potentials (EPSPs) were recorded using microelectrodes.
  • Fig. 4 shows the schematic of an EPSP recorded under normal conditions (A) , five minutes following replacement of 0 2 with N 2 (ischemic episode, B) , and 30 to 40 minutes following reoxygenation (C) .
  • the extent of permanent damage can be quantified by measuring both the amplitude (in mV) and the initial slope (in mV/msec) of the EPSP.
  • Figs. 5 and 6 show the protective effect of the antioxidant salen-Mn complex designated C7 in the rat brain slice ischemia EPSP system.
  • Brain slices were incubated in the absence or presence of 50 ⁇ M C7 and subjected to an episode of ischemia/reoxygenation. After 5 minutes of baseline recording, 0 2 was replaced by N 2 for an average of 5 minutes. 0 2 was then reintroduced and recording was continued for another 50 minutes. Samples with 50 ⁇ M C7 showed that both the amplitude and slopes of the EPSPs recovered to pre- ischemia levels. In contrast, recovery in untreated brain slices was only about 40% of pre-ischemia levels.
  • Fig. 7 demonstrates that, while without any treatment this percentage is very low (6%) , it was as high as 70% in slices treated with 50 ⁇ M C7. A slice was considered viable if an EPSP of 3 mV amplitude could be elicited by increasing stimulation intensity.
  • Tritiated mazindol was used for binding studies on samples of the globus pallidus, caudate nucleus, and striatum of mouse brain according to conventional methods; specific binding of tritiated mazindol was determined autoradiographically or by membrane binding (specific binding to the membrane fraction) . The experiment was performed over a 7 day period.
  • mice in the MPTP group were treated intraperitoneally with MPTP alone (40 mg/kg each day on days 1 and 2) .
  • Mice in the MPTP+C7 group were pretreated with C7 (33 mg/kg, i.p.) immediately prior to MPTP on days 1 and 2, and were given C7 (33 mg/kg) alone on day 3.
  • the animals were sacrificed after 7 days.
  • the results shown in Fig. 8 show a significant protective effect conferred in vivo by the salen- Mn complex, C7. Fig.
  • the perfusion fluid was a modified Krebs-Henseleit buffer containing (in mmol/1) : NaCl 118, KC1 5.9, NaHC0 3 25, MgCl 2 1.2, NaH 2 P0 4 0.6, CaCl 2 2.4, Glucose 11. pH was maintained at 7.4 ⁇ 0.05 when the perfusion medium was saturated with 0 2 -C0 2 (95%-5%) at 37°C.
  • the perfusion apparatus was fully thermostated such that the temperature of the perfusion medium was 37.0 ⁇ 0.5°C when it reached the aorta.
  • DMTU dimethylthiourea
  • Table V shows heart rates (HR) , systolic pressures (SP) , diastolic pressures (DP) , and the products HR x LVDP, in the three experimental groups, after 15 minutes of perfusion, before ischemia (Before) , 1 minute after reperfusion (1 After) and 15 minutes after reperfusion (15 After) .
  • the table also shows the number of hearts exhibiting episodes of ventricular fibrillation 1 minute after reperfusion (VF) .
  • Table VI summarizes the results from the electron microscopy evaluation of the hearts. Mitochondria were classified into Type A (normal) , Type B (swollen, unbroken) , and Type C (ruptured membranes) . Sarcomeres were classified into Type A (normal) and Type B (contacted and/or necrosis) . The results are expressed as percentages. The numbers of mitochondria analyzed were 1293, 1632 and 1595 for controls, DMTU and C7 groups, respectively. The numbers of sarcomeres analyzed were 1046, 1173, and 1143 for controls, DMTU and C7 groups, respectively. Sarcomeres Type A Type B
  • EAE is an animal model of multiple sclerosis. 30 SJL female mice, aged 10 weeks, were divided into 2 groups of 20 mice (control) and 10 mice (C7 treated) .
  • mice in both groups were immunized with an encephalitogenic PLP peptide in complete Freund's adjuvant subcutaneously, followed by Petrussis Toxin (IV) .
  • Petrussi ⁇ toxin was repeated on day 3 post immunization.
  • mice in the C7 group were treated daily (1 mg/mouse, approximately 40 mg/kg) by IP injection, starting from 2 days prior to immunization through day 14 after immunization.
  • mice in the control group developed symptomatic EAE: 2 Stage I, 4 Stage II/III, 2 Stage IV. During that same period, only one of 10 mice in the
  • Lipid peroxidation Hippocampal slices (400 ⁇ m thick) were obtained from
  • Sprague-Dawley rats (150-2OOg) and collected in preoxygenated (95% 0 2 / 5% C0 2 ) Krebs-Ringer phosphate medium (pH 7.4) containing NaCl 120 mM, KCl 5 mM, CaCl 2 1.3 mM, MgCl 2 1.2 mM, NaPhosphate 16 mM (pH 7.4) and glucose 10 mM.
  • the buffer was replaced with the same buffer (control) or a modified buffer (lactate buffer) containing NaCl 90 mM, KCl 5 mM, CaCl 2 1.3 mM, MgCl 2 1.2 mM, NaPhosphate 16 mM and lactic acid 30 mM (pH 5.0).
  • C7 50 ⁇ M was added during the preincubation and the incubation periods.
  • slices were collected and homogenized in 0.9 ml of TCA 5%, whereas 0.35 ml of TCA 5% was added to 0.5 ml of the incubation medium.
  • Lipid peroxidation was measured by adding 0.25 ml of a thiobarbituric acid reagent (TBAR) to 0.85 ml of the TCA extracts and incubating the mixture for 60 minutes at 85-93°C. Lipids were then extracted with 2 x 0.5 ml 1-butanol by vortexing for 10 seconds, then centrifuging at 2,000 rpm for 10 minutes. The absorbance of peroxidized lipids in the alcohol phase was measured in a spectrophotometer at 532 nm. Data were expressed as nmoles of malondialdehyde (MDA) using authentic MDA to establish a standard curve. Proteins were measured from an aliquot of the TCA extracts using the method of Bradford and the final results were calculated as nmoles MDA formed/mg protein.
  • MDA malondialdehyde
  • Hippocampal slices were collected in preoxygenated Krebs-Ringer phosphate buffer, with or without 50 ⁇ M C7, at 35°C in a shaking water bath.
  • slices were transferred into the same buffer or in a buffer containing 30 mM lactate, pH 5.0 (with or without C7) .
  • Slices from all groups were collected after a 100 minute incubation and tested for lipid peroxidation, as indicated by malondialdehyde reaction with thiobarbituric acid.
  • MPTP in mice were administered two injections of MPTP dissolved in normal saline (40 mg/kg, s.c.) 24 hours apart. A group of animals also received C7 in three injections (33 mg/kg, s.c.) administered 24 hours apart, starting 1 day before the onset of MPTP treatment. Animals were sacrificed 7 days after the first MPTP injection, and neuronal pathology was assessed by the binding of 3 H-mazindol, a ligand for the dopamine transporter, to 10 mm frozen brain sections or to striatal homogenates.
  • 6-0HDA in mice.
  • Adult male CFW mice were anesthetized with ketamine and rumpun, and immobilized in a stereotaxic device.
  • 6-0HDA as the hydrobromide salt, was dissolved in normal saline with 1% ascorbate, and 50 ⁇ g was administered in lateral ventricle by means of a 10 ⁇ l Hamilton syringe.
  • C7 (66 mg/kg, i.p.) was administered daily for 4 days. Animals were sacrificed 7 days later, and neuronal pathology was assessed by measuring 3 H-mazindol binding in striatal homogenates.
  • RESULTS C7 protects hippocampal slices from anoxia-induced damage
  • C7 provided a significant degree of protection against anoxia-induced decrease in synaptic response in CA1.
  • the decrease in both EPSP slope (A) and amplitude (B) were prevented by C7.
  • Purified bovine SOD in the same assay provided no protection) .
  • Fig. lo shows I.e.v. injection of 6-OHDA (50 ⁇ g) resulted in a 60-70% decrease in mazindol binding in homogenates from the striatum ipsilateral from the injection site and a 30% decrease from the contralateral striatum (Fig. 10) .
  • Treatment with C7 (4x66 mg/kg) produced a significant reduction in the ipsilateral side and a complete protection in the contralateral side.
  • C7 Synthetic Catalytic Scavenger
  • antioxidant salen-metal complexess such as C7 might have a wide range of therapeutic applications.

Abstract

L'invention se rapporte à des complexes salen-métal antioxydants se présentant sous une forme appropriée pour être administrés comme produits pharmaceutiques afin de traiter ou prévenir une maladie associée à des lésions cellulaires ou tissulaires produites par des radicaux libres tels que le superoxyde.
PCT/US1993/011857 1992-12-07 1993-12-06 Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies WO1994013300A1 (fr)

Priority Applications (19)

Application Number Priority Date Filing Date Title
AT94903498T ATE290867T1 (de) 1992-12-07 1993-12-06 Synthetische katalytische freie radikalfänger, verwendbar als antioxidantien zur prävention und therapie von krankheiten
GB9415050A GB2277873B (en) 1992-12-07 1993-12-06 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
UA95062707A UA27949C2 (uk) 1992-12-07 1993-12-06 Фармацевтична композиція для запобігання або терапії захворювань, які пов`язані з вільними радикалами і які є результатом дисбалансу вільних радикалів (варіанти), спосіб запобігання, зупинки або лікування захворювань, які пов`язані з вільними радикалами і які є результатом дисбалансу вільних радикалів, композиція для запобігання пов`язаних з вільними радикалами променевих або хімічних уражень пацієнта, спосіб запобігання, зупинки або лікування неврологічних уражень, які викликані 1-метил-4-феніл-1,2,3,6-тетрагідропіридином (мртр) або гіпоксичними ураженнями
CA002150937A CA2150937C (fr) 1992-12-07 1993-12-06 Agents catalytiques synthetiques, suppresseurs de radicaux libres, utiles comme antioxydants pour la prevention et le traitement de maladies
SK745-95A SK74595A3 (en) 1992-12-07 1993-12-06 Synthetic catalytic free radical scavengers usefull as antioxidants for preventions and therapy of disease
EP94903498A EP0746321B1 (fr) 1992-12-07 1993-12-06 Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies
HU9501644A HU225109B1 (en) 1992-12-07 1993-12-06 Pharmaceutical compositions comprising antioxidant salen-metal complex and their use
PL93309334A PL175446B1 (pl) 1992-12-07 1993-12-06 Kompozycja farmaceutyczna do zapobiegania lub leczenia stanów chorobowych związanych z wolnymi rodnikami
JP51432894A JP3502099B2 (ja) 1992-12-07 1993-12-06 疾患の予防および治療のための酸化防止剤として有用な合成触媒のフリーラジカルスカベンジャー
AU57419/94A AU697399B2 (en) 1992-12-07 1993-12-06 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
KR10-2003-7004595A KR20040004386A (ko) 1992-12-07 1993-12-06 질병의 예방 및 치료용 항산화제로서 유용한 합성 촉매자유 라디칼 스캐빈저
DE69333775T DE69333775T2 (de) 1992-12-07 1993-12-06 Synthetische katalytische freie radikalfänger, verwendbar als antioxidantien zur prävention und therapie von krankheiten
LVP-95-158A LV10924B (en) 1992-12-07 1995-06-06 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
NO19952237A NO319454B1 (no) 1992-12-07 1995-06-06 Farmasoytiske preparater basert pa syntetiske katalytiske fri-radikal-oppfangere anvendelige som anti-oksidanter for forebyggelse og terapi av sykdom
US08/479,697 US5834509A (en) 1992-12-07 1995-06-07 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US08/485,489 US5696109A (en) 1992-12-07 1995-06-07 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US09/542,182 US6573257B2 (en) 1993-12-06 2000-04-04 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US10/452,695 US6900198B2 (en) 1993-12-06 2003-05-30 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US11/362,454 US7582786B2 (en) 1992-12-07 2006-02-23 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/987,474 US5403834A (en) 1992-12-07 1992-12-07 Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US07/987,474 1992-12-07

Publications (2)

Publication Number Publication Date
WO1994013300A1 true WO1994013300A1 (fr) 1994-06-23
WO1994013300A9 WO1994013300A9 (fr) 1994-08-18

Family

ID=25533292

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/011857 WO1994013300A1 (fr) 1992-12-07 1993-12-06 Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies

Country Status (20)

Country Link
US (2) US5403834A (fr)
EP (2) EP1642583A2 (fr)
JP (1) JP3502099B2 (fr)
KR (1) KR20040004386A (fr)
AT (1) ATE290867T1 (fr)
AU (1) AU697399B2 (fr)
CA (1) CA2150937C (fr)
CZ (1) CZ296288B6 (fr)
DE (1) DE69333775T2 (fr)
GB (2) GB2277873B (fr)
HU (1) HU225109B1 (fr)
LV (1) LV10924B (fr)
NO (1) NO319454B1 (fr)
NZ (1) NZ259200A (fr)
OA (1) OA10165A (fr)
PL (1) PL175446B1 (fr)
RU (1) RU2157213C2 (fr)
SK (1) SK74595A3 (fr)
UA (1) UA27949C2 (fr)
WO (1) WO1994013300A1 (fr)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995031197A1 (fr) * 1994-05-13 1995-11-23 Monsanto Company Procedes d'utilisation de catalyseurs de decomposition deperoxynitrite et compositions pharmaceutiques a cet effet
EP0831836A1 (fr) * 1995-06-07 1998-04-01 Eukarion, Inc. Intercepteurs catalytiques synthetiques de radicaux libres s'utilisant comme antioxydants dans la prevention et le traitement de maladies
US6245758B1 (en) 1994-05-13 2001-06-12 Michael K. Stern Methods of use for peroxynitrite decomposition catalysts, pharmaceutical compositions therefor
US6541490B1 (en) 1998-08-17 2003-04-01 Eukarion, Inc. Bipyridine manganese complexes
US7582786B2 (en) 1992-12-07 2009-09-01 Eukarion Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US7820788B2 (en) 2002-12-09 2010-10-26 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
EP2357166A1 (fr) * 2008-11-20 2011-08-17 IHI Corporation Composé complexe de métal-salène auto-magnétique
EP2564852A1 (fr) * 2010-04-28 2013-03-06 IHI Corporation Médicament traitant les tumeurs cérébrales
EP2738158A1 (fr) * 2011-06-13 2014-06-04 IHI Corporation Composé complexe métal salène, anesthésique local, et agent tumoral anti-malignité
EP2772522A4 (fr) * 2011-10-27 2016-02-17 Ihi Corp Inhibiteur de radicaux
US9468621B2 (en) 2011-10-04 2016-10-18 Ihi Corporation Metal-salen complex compound responsive drug and intra-corporeal behavior control system for metal-salen complex compound
US9592219B2 (en) 2011-07-26 2017-03-14 Ihi Corporation Self-magnetic metal-salen complex compound
CN108030926A (zh) * 2017-12-15 2018-05-15 武汉工程大学 一种具有高的SOD酶活性的Salen型锰配合物-壳聚糖复合材料及其制备方法
US10034941B2 (en) 2007-12-28 2018-07-31 Ihi Corporation Iron-salen complex

Families Citing this family (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE36460E (en) * 1990-10-10 1999-12-21 Life Science Holdings, Inc. Method of providing circulation via lung expansion and deflation
US5653685A (en) * 1990-10-10 1997-08-05 Lrt, Inc. Method of providing circulation via lung expansion and deflation
US5834509A (en) * 1992-12-07 1998-11-10 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US5403834A (en) * 1992-12-07 1995-04-04 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US5686436A (en) * 1993-05-13 1997-11-11 Hiv Diagnostics, Inc. Multi-faceted method to repress reproduction of latent viruses in humans and animals
US6127356A (en) 1993-10-15 2000-10-03 Duke University Oxidant scavengers
US5681278A (en) * 1994-06-23 1997-10-28 Cormedics Corp. Coronary vasculature treatment method
WO1996004311A1 (fr) * 1994-08-03 1996-02-15 University Of Alabama At Birmingham Research Foundation Anticorps monoclonal dirige contre la nitrotyrosine, procedes de diagnostic et procedes therapeutiques
US5900433A (en) * 1995-06-23 1999-05-04 Cormedics Corp. Vascular treatment method and apparatus
CA2247876A1 (fr) * 1996-03-08 1997-09-12 Ronald M. Klatz Procede et dispositif de ventilation liquide
WO1998010057A1 (fr) 1996-09-06 1998-03-12 Emory University Methodes d'essai de composes destines a etre utilises en tant qu'antioxydants therapeutiques
US6369106B1 (en) * 1996-12-26 2002-04-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Treatment of ischemic brain injuries with brain targeted anti oxidant compounds
US6214817B1 (en) 1997-06-20 2001-04-10 Monsanto Company Substituted pyridino pentaazamacrocyle complexes having superoxide dismutase activity
US5834178C1 (en) * 1997-07-09 2002-04-23 Univ Wayne State Flush-storage solution for donor organs
US5829449A (en) * 1997-09-19 1998-11-03 Thione International, Inc. Smoking products containing antioxidants
AT409306B (de) * 1997-10-03 2002-07-25 Hoffmann La Roche Optisch chemischer sensor
CA2309154C (fr) 1997-11-03 2010-02-16 Duke University Porphyrines substituees
CA2249778A1 (fr) * 1997-12-15 1999-06-15 Bayer Corporation Dosage competitif d'apo-peroxydase
US7816403B2 (en) * 1998-09-08 2010-10-19 University Of Utah Research Foundation Method of inhibiting ATF/CREB and cancer cell growth and pharmaceutical compositions for same
US6703384B2 (en) * 1998-09-23 2004-03-09 Research Development Foundation Tocopherols, tocotrienols, other chroman and side chain derivatives and uses thereof
KR100753184B1 (ko) 1999-01-25 2007-08-30 내셔날 쥬이쉬 메디칼 앤드 리서치 센터 치환된 포르피린
US6365385B1 (en) 1999-03-22 2002-04-02 Duke University Methods of culturing and encapsulating pancreatic islet cells
US6303355B1 (en) 1999-03-22 2001-10-16 Duke University Method of culturing, cryopreserving and encapsulating pancreatic islet cells
JP2003500174A (ja) 1999-05-27 2003-01-07 フアルマシア・コーポレーシヨン スーパーオキシド・ジスムターゼ模倣体で修飾された生体材料
US20040110722A1 (en) * 1999-05-27 2004-06-10 Ornberg Richard L. Modified hyaluronic acid polymers
US6179804B1 (en) * 1999-08-18 2001-01-30 Oxypatch, Llc Treatment apparatus for wounds
RU2263672C2 (ru) * 2000-02-11 2005-11-10 Рисерч Дивелопмент Фаундейшн Токоферолы, токотриенолы, другие хроманы и производные по боковым цепям и их применение
US20040047852A1 (en) * 2001-03-02 2004-03-11 Kennedy Thomas Preston Method of treating cancer
US20030069281A1 (en) * 2000-06-14 2003-04-10 Irwin Fridovich Tetrapyrroles
US6589948B1 (en) 2000-11-28 2003-07-08 Eukarion, Inc. Cyclic salen-metal compounds: reactive oxygen species scavengers useful as antioxidants in the treatment and prevention of diseases
US6481442B1 (en) 2000-11-28 2002-11-19 Lorillard Licensing Company, Llc Smoking article including a filter for selectively removing carbonyls
WO2002043514A1 (fr) * 2000-11-28 2002-06-06 Lorillard Licensing Company, Llc Article pour fumeur comportant une pompe sélective à co
US6692458B2 (en) * 2000-12-19 2004-02-17 Edwards Lifesciences Corporation Intra-pericardial drug delivery device with multiple balloons and method for angiogenesis
DE60233317D1 (de) * 2001-01-19 2009-09-24 Nat Jewish Med & Res Center Medikament zum schutz in der radiotherapie
ITPI20010014A1 (it) * 2001-03-05 2002-09-05 Ivo Pera Composto per filtri per sigarette,o altri articoli da fumo,a base di sostanze antiossidanti ed il filtro cosi'ottenuto
KR100681326B1 (ko) * 2001-04-20 2007-02-15 주식회사 중외제약 신규한 망간-키랄성 화합물 및 그의 이용방법
JP2005508864A (ja) * 2001-06-01 2005-04-07 ナショナル・ジュウィッシュ・メディカル・アンド・リサーチ・センター 糖尿病の治療、または移植術における使用、または免疫寛容の誘導のためのオキシダントスカベンジャー
WO2003010153A1 (fr) * 2001-07-26 2003-02-06 Samsung Electronics Co. Ltd. Composes de n-alkyl-n-phenylhydroxylamine contenant des groupes chelateurs de metaux, leur preparation et leurs applications therapeutiques
WO2003010143A1 (fr) 2001-07-26 2003-02-06 Samsung Electronics Co., Ltd. Derives de l'acide dialkylhydroxybenzoique contenant des groupes chelateurs de metaux et leurs utilisations therapeutiques
WO2003010154A1 (fr) 2001-07-26 2003-02-06 Samsung Electronics Co. Ltd. Composes seleno contenant un groupe fonctionnel nitron, leur preparation et leurs applications therapeutiques
US20030236301A1 (en) * 2001-12-19 2003-12-25 Bob Sanders Liposomal delivery of vitamin E based compounds
DE60236456D1 (de) * 2001-12-19 2010-07-01 Res Dev Foundation Liposomale abgabe von verbindungen auf vitamin-e-basis
JP2006501163A (ja) * 2002-06-07 2006-01-12 デューク・ユニバーシティー 置換ポルフィリン
US6835387B2 (en) * 2002-06-11 2004-12-28 Scimed Life Systems, Inc. Sustained release of superoxide dismutase mimics from implantable or insertable medical devices
WO2004017959A2 (fr) * 2002-08-22 2004-03-04 University Of Florida Activite antioxydante et de piegeage de radicaux d'analogues synthetiques de desferrithiocine
US20040044220A1 (en) * 2002-08-22 2004-03-04 University Of Florida Antioxidant and radical scavenging activity of synthetic analogs of desferrithiocin
RU2361615C2 (ru) * 2002-12-09 2009-07-20 Абраксис Байосайенс, Ллс. Композиции и способы доставки фармакологических агентов
AU2003302898A1 (en) * 2002-12-11 2004-06-30 Cryolife, Inc. Radical retardant cryopreservation solutions
WO2005030137A2 (fr) * 2003-09-24 2005-04-07 The University Of Toledo, A University Instrumentality Of The State Of Ohio Procede ameliorer la performance biomecanique de matieres biologiques irradiees et matieres formees au moyen de ce procede
US20050100537A1 (en) * 2003-11-10 2005-05-12 Evans Gregory S. Methods and kits for reducing cellular damage, inhibiting free radical production, and scavenging free radicals in mammals
US20090099150A1 (en) * 2005-01-19 2009-04-16 Daniela Salvemini Methotrexate Combinations For Treating Inflammatory Diseases
KR101159392B1 (ko) * 2005-05-04 2012-06-28 코티 프레스티지 랭카스터 그룹 게엠베하 화학요법에 의해 야기되는 피부 및 모발의 손상을 예방또는 치료하기 위한 용도를 지닌 유리-라디칼 스캐빈저
US8246975B2 (en) * 2006-06-28 2012-08-21 Ihi Corporation Drug, drug guidance system, magnetic detection system, and drug design method
US8252780B2 (en) * 2006-12-01 2012-08-28 Women & Infants' Hospital Organometallic complexes as therapeutic agents
ES2600469T3 (es) 2008-05-23 2017-02-09 National Jewish Health Un compuesto para su uso en el tratamiento de lesiones asociadas con la exposición al gas fosgeno o al gas de cloro
KR101050273B1 (ko) * 2008-11-21 2011-07-19 세종대학교산학협력단 살렌 전이금속 착화합물, 및 그 제조 방법
CN102834512A (zh) * 2010-04-06 2012-12-19 株式会社Ihi 金属salen配合物衍生物及其制造方法
WO2011151978A1 (fr) * 2010-06-01 2011-12-08 株式会社Ihi Matériau colorant fluorescent et son utilisation
WO2012086683A1 (fr) * 2010-12-21 2012-06-28 株式会社Ihi Composé complexe métal-salen et son procédé de production
RU2622646C2 (ru) * 2012-01-05 2017-06-19 Пледфарма Аб Калмангафодипир, новое химическое соединение и другие смешанные комплексные соединения с металлами, способы получения, композиции и способы лечения
RU2699040C1 (ru) * 2018-07-23 2019-09-03 Федеральное государственное бюджетное учреждение науки "Уральский научно-практический центр радиационной медицины Федерального медико-биологического агентства" (ФГБУН УНПЦ РМ ФМБА России) Способ экстренной профилактики и лечения острой лучевой болезни (варианты)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4540573A (en) * 1983-07-14 1985-09-10 New York Blood Center, Inc. Undenatured virus-free biologically active protein derivatives

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5106841A (en) * 1986-05-13 1992-04-21 Chai-Tech Corporation Antiviral compositions and method for their use
US5223538A (en) * 1987-03-31 1993-06-29 Duke University Superoxide dismutase mimic
EP0643626B1 (fr) * 1991-08-26 2002-02-27 Research Corporation Technologies, Inc Procede de production d'epoxychromannes avec un catalyseur chiral
US5403834A (en) * 1992-12-07 1995-04-04 Eukarion, Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4540573A (en) * 1983-07-14 1985-09-10 New York Blood Center, Inc. Undenatured virus-free biologically active protein derivatives

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SITTIG M: "HANDBOOK OF TOXIC AND HAZARDOUS CHEMICALS AND CARCINOGENS, PASSAGE", HANDBOOK OF TOXIC AND HAZARDOUS CHEMICALS AND CARCINOGENS, XX, XX, no. 02, 1 January 1985 (1985-01-01), XX, pages 638 - 641 + 559, XP002945167 *
WOLFF M. E.: "BURGER'S MEDICINAL CHEMISTRY. BASIS OF MEDICINAL CHEMISTRY.", vol. ED. 4, 1 January 1979, NEW YORK, WILEY & SONS., US, article "BURGER'S MEDICINAL CHEMISTRY", pages: 10/11 + 22 + 29 - 35 + 39, XP002945123, 017014 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7582786B2 (en) 1992-12-07 2009-09-01 Eukarion Inc. Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
WO1995031197A1 (fr) * 1994-05-13 1995-11-23 Monsanto Company Procedes d'utilisation de catalyseurs de decomposition deperoxynitrite et compositions pharmaceutiques a cet effet
US6245758B1 (en) 1994-05-13 2001-06-12 Michael K. Stern Methods of use for peroxynitrite decomposition catalysts, pharmaceutical compositions therefor
EP0831836A1 (fr) * 1995-06-07 1998-04-01 Eukarion, Inc. Intercepteurs catalytiques synthetiques de radicaux libres s'utilisant comme antioxydants dans la prevention et le traitement de maladies
EP0831836A4 (fr) * 1995-06-07 2000-09-27 Eukarion Inc Intercepteurs catalytiques synthetiques de radicaux libres s'utilisant comme antioxydants dans la prevention et le traitement de maladies
US6541490B1 (en) 1998-08-17 2003-04-01 Eukarion, Inc. Bipyridine manganese complexes
US8846771B2 (en) 2002-12-09 2014-09-30 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US7820788B2 (en) 2002-12-09 2010-10-26 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US9012518B2 (en) 2002-12-09 2015-04-21 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US9012519B2 (en) 2002-12-09 2015-04-21 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US10034941B2 (en) 2007-12-28 2018-07-31 Ihi Corporation Iron-salen complex
US9505732B2 (en) 2008-11-20 2016-11-29 Ihi Corporation Auto magnetic metal salen complex compound
EP2357166A4 (fr) * 2008-11-20 2012-07-04 Ihi Corp Composé complexe de métal-salène auto-magnétique
EP2357166A1 (fr) * 2008-11-20 2011-08-17 IHI Corporation Composé complexe de métal-salène auto-magnétique
EP2564852A1 (fr) * 2010-04-28 2013-03-06 IHI Corporation Médicament traitant les tumeurs cérébrales
EP2564852A4 (fr) * 2010-04-28 2013-10-02 Ihi Corp Médicament traitant les tumeurs cérébrales
US8933118B2 (en) 2010-04-28 2015-01-13 Ihi Corporation Anti-brain-tumor drug
EP2738158A1 (fr) * 2011-06-13 2014-06-04 IHI Corporation Composé complexe métal salène, anesthésique local, et agent tumoral anti-malignité
EP2738158A4 (fr) * 2011-06-13 2014-12-17 Ihi Corp Composé complexe métal salène, anesthésique local, et agent tumoral anti-malignité
US10034851B2 (en) 2011-06-13 2018-07-31 Ihi Corporation Metal-salen complex compound, local anesthetic and antineoplastic drug
US9592219B2 (en) 2011-07-26 2017-03-14 Ihi Corporation Self-magnetic metal-salen complex compound
US9468621B2 (en) 2011-10-04 2016-10-18 Ihi Corporation Metal-salen complex compound responsive drug and intra-corporeal behavior control system for metal-salen complex compound
US9434880B2 (en) 2011-10-27 2016-09-06 Ihi Corporation Radical inhibitor
EP2772522A4 (fr) * 2011-10-27 2016-02-17 Ihi Corp Inhibiteur de radicaux
CN108030926A (zh) * 2017-12-15 2018-05-15 武汉工程大学 一种具有高的SOD酶活性的Salen型锰配合物-壳聚糖复合材料及其制备方法
CN108030926B (zh) * 2017-12-15 2021-07-13 武汉工程大学 一种具有高的SOD酶活性的Salen型锰配合物-壳聚糖复合材料及其制备方法

Also Published As

Publication number Publication date
AU697399B2 (en) 1998-10-08
GB2305107B (en) 1997-05-14
OA10165A (en) 1996-12-18
CZ296288B6 (cs) 2006-02-15
NO952237L (no) 1995-08-01
GB2277873A (en) 1994-11-16
PL309334A1 (en) 1995-10-02
LV10924A (lv) 1995-12-20
JPH08504211A (ja) 1996-05-07
US5827880A (en) 1998-10-27
KR20040004386A (ko) 2004-01-13
NO319454B1 (no) 2005-08-15
GB2305107A (en) 1997-04-02
HU225109B1 (en) 2006-06-28
ATE290867T1 (de) 2005-04-15
GB9621087D0 (en) 1996-11-27
AU5741994A (en) 1994-07-04
CA2150937A1 (fr) 1994-06-23
UA27949C2 (uk) 2000-10-16
JP3502099B2 (ja) 2004-03-02
RU2157213C2 (ru) 2000-10-10
EP0746321A4 (fr) 2001-05-09
DE69333775T2 (de) 2006-04-06
SK74595A3 (en) 1996-05-08
EP0746321A1 (fr) 1996-12-11
EP1642583A2 (fr) 2006-04-05
CA2150937C (fr) 2007-08-14
NO952237D0 (no) 1995-06-06
LV10924B (en) 1996-10-20
NZ259200A (en) 1997-06-24
CZ144295A3 (en) 1996-10-16
PL175446B1 (pl) 1998-12-31
HUT72967A (en) 1996-06-28
GB2277873B (en) 1997-05-14
GB9415050D0 (en) 1994-09-14
EP0746321B1 (fr) 2005-03-16
US5403834A (en) 1995-04-04
DE69333775D1 (de) 2005-04-21
HU9501644D0 (en) 1995-08-28

Similar Documents

Publication Publication Date Title
AU697399B2 (en) Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
WO1994013300A9 (fr) Piegeurs de radicaux libres catalytiques synthetiques utiles comme antioxydants dans la prevention et la therapie de maladies
US6900198B2 (en) Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US6589948B1 (en) Cyclic salen-metal compounds: reactive oxygen species scavengers useful as antioxidants in the treatment and prevention of diseases
US7582786B2 (en) Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
US5834509A (en) Synthetic catalytic free radical scavengers useful as antioxidants for prevention and therapy of disease
AU2002239381A1 (en) Cyclic salen-metal compounds as scavengers for oxygen radicals and useful as antioxidants in the treatment and prevention of diseases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR BY CA CH CZ DE DK ES FI GB HU JP KP KR KZ LK LU LV MG MN MW NL NO NZ PL PT RO RU SD SE SK UA US VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 9/11 AND 10/11,DRAWINGS,REPLACED BY NEW PAGES BEARING THE SAME NUMBER;DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2150937

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 74595

Country of ref document: SK

Ref document number: PV1995-1442

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 259200

Country of ref document: NZ

Ref document number: 1019950702375

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1994903498

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV1995-1442

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1994903498

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020037004595

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1994903498

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: PV1995-1442

Country of ref document: CZ